CN115322238A - Oxysterol and methods of use thereof - Google Patents

Oxysterol and methods of use thereof Download PDF

Info

Publication number
CN115322238A
CN115322238A CN202210876413.0A CN202210876413A CN115322238A CN 115322238 A CN115322238 A CN 115322238A CN 202210876413 A CN202210876413 A CN 202210876413A CN 115322238 A CN115322238 A CN 115322238A
Authority
CN
China
Prior art keywords
substituted
alkyl
unsubstituted
hydrogen
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210876413.0A
Other languages
Chinese (zh)
Inventor
F.G.萨利图罗
A.J.罗比乔德
G.马蒂内兹博特拉
B.L.哈里森
A.格里芬
D.拉
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sage Therapeutics Inc
Original Assignee
Sage Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sage Therapeutics Inc filed Critical Sage Therapeutics Inc
Publication of CN115322238A publication Critical patent/CN115322238A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J17/00Normal steroids containing carbon, hydrogen, halogen or oxygen, having an oxygen-containing hetero ring not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J43/00Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • C07J43/003Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton not condensed
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • C07J9/005Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane containing a carboxylic function directly attached or attached by a chain containing only carbon atoms to the cyclopenta[a]hydrophenanthrene skeleton

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Immunology (AREA)
  • Pain & Pain Management (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Rheumatology (AREA)
  • Psychiatry (AREA)
  • Dermatology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Anesthesiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychology (AREA)
  • Transplantation (AREA)
  • Epidemiology (AREA)

Abstract

Providing a compound according to formula (a), and pharmaceutically acceptable salts thereof and pharmaceutical compositions thereof; wherein R is 1 、R 2 、R 3 、R 4 、R 5 、R 6 And R G As defined herein. The compounds of the present invention are expected to be useful in the prevention and treatment of a variety of conditions.

Description

Oxysterol and methods of use thereof
The application is based on the application date of 2017, 10, 18 and the application number of 201780076791.8 (PCT/US 2017/057276), and the invention name is as follows: divisional application of the patent application for oxysterol and methods of use thereof.
Cross Reference to Related Applications
The present application claims priority to U.S. provisional application No. 62/409,756, filed on 18/10/2016 and U.S. provisional application No. 62/409,768, filed on 18/10/2016, each of which is incorporated herein by reference in its entirety.
Background
NMDA receptors are heteromeric complexes composed of NR1, NR2 and/or NR3 subunits, and have distinct recognition sites for exogenous and endogenous ligands. These recognition sites include binding sites for glycine as well as binding sites for glutamate agonists and modulators. NMDA receptors are expressed in peripheral tissues and CNS, where they are involved in excitatory synaptic transmission. Activation of these receptors contributes to synaptic plasticity in some cases and leads to excitotoxicity in other cases. These receptors are ligands Body-gated ion channels that allow Ca upon binding of glutamate and glycine 2+ Through, and are important for, excitatory neurotransmission and normal CNS function. Positive modulators may be useful as therapeutic agents with potential clinical use as cognitive enhancers and in the treatment of psychotic disorders in which glutamatergic transmission is reduced or defective (see for example Horak et al, j. Of Neuroscience,2004, 24 (46), 10318-10325). Conversely, negative modulators may be useful as therapeutic agents, having potential clinical use in the treatment of psychiatric disorders in which glutamatergic transmission is pathologically increased (e.g., in the treatment of refractory depression).
Oxysterol (Oxysterol) is a cholesterol analog that is a modulator of NMDA receptor function. There is a need for new oxysterol that modulate NMDA receptors to prevent and treat conditions associated with NMDA expression and function. The compounds, compositions, and methods described herein are useful for these purposes.
Disclosure of Invention
Provided herein are substituted oxysterol useful for preventing and/or treating various diseases (including but not limited to NMDA-mediated diseases). Also provided are pharmaceutical compositions comprising the compounds of the invention, and methods of use and treatment thereof.
In one aspect, provided herein is a compound according to formula (a), or a pharmaceutically acceptable salt thereof:
Figure BDA0003762336660000021
or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen or alkyl (e.g. C) 1 -C 6 Alkyl groups); r 2 And R 3 Each independently hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, or heteroaryl, or R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring; r 4 And R 5 Each independently is hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl radical(e.g., C) 1 -C 3 Alkyl) or R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group; r 6 Absent or hydrogen; r is G Is hydrogen or alkyl; and is provided with
Figure BDA0003762336660000022
Represents a single or double bond, wherein
Figure BDA0003762336660000023
When one is a double bond, the other is
Figure BDA0003762336660000024
Is a single bond and R 6 Is absent; and when two are
Figure BDA0003762336660000025
When all are single bonds, then R 6 Is hydrogen.
In some embodiments, R 1 Is alkyl (e.g. C) 1 -C 6 Alkyl groups). In some embodiments, R 1 Is C 1 -C 6 Alkyl (e.g., -CH) 3 、–CH 2 CH 3 、–CH 2 OCH 3 or-CF 3 ). In some embodiments, R 1 is-CH 3 、–CF 3 or-CH 2 CH 3 . In some embodiments, R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl).
In some embodiments, R 2 Is hydrogen or alkyl (e.g. C) 1 -C 6 Alkyl groups).
In some embodiments, R 2 And R 3 Each independently hydrogen or alkyl (e.g., C) 1 -C 6 Alkyl groups). In some embodiments, R 2 And R 3 Each independently is hydrogen or C 1 -C 6 Haloalkyl (e.g., -CF) 3 ). In some embodiments, R 2 And R 3 Are each independently hydrogen-CF 3 or-CH 3
In some embodiments, R 4 is-OH or halogen (e.g., -F).
In some embodiments, R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group.
In some embodiments, R 4 Is hydrogen and R 5 Is halogen (e.g., -F). In some embodiments, R 4 And R 5 Is halogen (e.g., -F). In some embodiments, R 4 And R 5 Is hydrogen.
In some embodiments, R 2 Is aryl or heteroaryl and R 3 Is hydrogen. In some embodiments, R 2 Is carbocyclyl or heterocyclyl and R 3 Is hydrogen. In some embodiments, R 2 And R 3 Is hydrogen. In some embodiments, R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered carbocyclic or heterocyclic ring.
In some embodiments, R 6 Is hydrogen and
Figure BDA0003762336660000031
represents a single bond.
In some embodiments, R G Is hydrogen or-CH 3
In one aspect, provided herein are compounds according to formula (I-63):
Figure BDA0003762336660000032
or a pharmaceutically acceptable salt thereof, wherein: r 1 Is alkyl (e.g. C) 1 -C 6 Alkyl groups); r 2 And R 3 Each independently hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl or heteroaryl, or R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring; r 4 And R 5 Each independently is hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl) or R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group; r is 6 Absent or hydrogen; and is
Figure BDA0003762336660000033
Represents a single or double bond, wherein
Figure BDA0003762336660000034
When one is a double bond, the other is
Figure BDA0003762336660000035
Is a single bond and R 6 Is absent; and when two are
Figure BDA0003762336660000036
When all are single bonds, then R 6 Is hydrogen.
In some embodiments, R 1 Is alkyl (e.g. C) 1 -C 6 Alkyl). In some embodiments, R 1 Is C 1 -C 6 Alkyl (e.g., -CH) 3 、–CH 2 CH 3 、–CH 2 OCH 3 or-CF 3 ). In some embodiments, R 1 is-CH 3 、–CF 3 or-CH 2 CH 3 . In some embodiments, R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl groups). In some embodiments, R 1 Is an unsubstituted alkyl group. In some embodiments R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 Alkyl (e.g., -CH) 3 )。
In some embodiments, R 2 Is alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl and R 3 Is hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), alkenyl, alkynyl, carbocycleRadical, heterocyclyl radical, aryl or heteroaryl radical or R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered ring.
In some embodiments, R 2 Is alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl and R 3 Is hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl or R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered ring.
In some embodiments, R 2 Is hydrogen or alkyl (e.g. C) 1 -C 6 Alkyl groups).
In some embodiments, R 2 And R 3 Each independently hydrogen or alkyl (e.g., C) 1 -C 6 Alkyl groups). In some embodiments, R 2 And R 3 Each independently is hydrogen or C 1 -C 6 Haloalkyl (e.g., -CF) 3 ). In some embodiments, R 2 And R 3 Each independently is C 5 Alkyl (e.g., substituted or unsubstituted isoamyl) or hydrogen. In some embodiments, R 2 And R 3 Each independently is an isoamyl group (e.g., a substituted or unsubstituted isoamyl group) or hydrogen. In some embodiments, R 2 And R 3 Each independently is hydrogen, -CF 3 or-CH 3
In some embodiments, R 4 is-OH or halogen (e.g., -F).
In some embodiments, R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group. In some embodiments, R 4 Is hydrogen and R 5 Is halogen (e.g., -F).
In some embodiments, R 4 And R 5 Is halogen (e.g., -F). In some embodiments, R 4 And R 5 Is hydrogen.
In some embodiments, R 2 Is aryl or heteroaryl and R 3 Is hydrogen. In some embodiments, R 2 Is a carbocyclic ringOr a heterocyclic radical and R 3 Is hydrogen. In some embodiments, R 2 And R 3 Is hydrogen. In some embodiments, R 2 Is an isoamyl group (e.g., substituted or unsubstituted isoamyl group) and R 3 Is hydrogen. In some embodiments, R 2 is-CF 3 or-CH 3 And R is 3 Is hydrogen or-CH 3 . In some embodiments, R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered carbocyclic or heterocyclic ring.
In some embodiments, R 1 is-CH 3 or-CH 2 CH 3 ,R 2 Is an isoamyl group (e.g., substituted or unsubstituted isoamyl group), and R 3 Is hydrogen. In some embodiments, R 1 is-CH 3 or-CH 2 CH 3 ,R 2 Is unsubstituted isopentyl, and R 3 Is hydrogen.
In some embodiments, R 2 Is unsubstituted C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group. In some embodiments, R 2 Is unsubstituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is a pyridyl group. In some embodiments, each R is 2 Is isopentyl and R 3 Is hydrogen. In some embodiments, R 2 is-CF 3 And R is 3 Is hydrogen. In some embodiments, R 2 Is unsubstituted alkyl (e.g., unsubstituted C) 1 -C 6 Alkyl groups). In some embodiments, R 2 Is a carbocyclylalkyl group. In some embodiments, R 2 Is carbocyclylalkyl and R 3 Is hydrogen. In some embodiments, R 2 Is an aralkyl group (e.g., benzyl). In some embodiments, R 2 Is a heterocyclylalkyl group. In some embodiments, wherein R is 2 Is unsubstituted C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, carbocyclyl, carbocyclylalkyl, aralkyl, or heterocyclylalkyl.
In some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-A63), (I-B63), or (I-C63):
Figure BDA0003762336660000051
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-A63):
Figure BDA0003762336660000052
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-C63):
Figure BDA0003762336660000053
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-B63):
Figure BDA0003762336660000054
in some embodiments, the compound of formula (I63) is selected from compounds of formula (I-D63):
Figure BDA0003762336660000061
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-E63):
Figure BDA0003762336660000062
In some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-D-I63) or (I-D-ii 63):
Figure BDA0003762336660000063
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-E-I63) or (I-E-ii 63):
Figure BDA0003762336660000064
Figure BDA0003762336660000071
in one aspect, provided herein are compounds according to formula (I-67):
Figure BDA0003762336660000072
or a pharmaceutically acceptable salt thereof, wherein: r 1 Is hydrogen or alkyl (e.g. C) 1 -C 6 Alkyl groups); r 2 And R 3 Each independently hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl, or R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring; r 4 And R 5 Each independently is hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl), or R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group; r 6 Absent or hydrogen; and is provided with
Figure BDA0003762336660000073
Represents a single or double bond, wherein
Figure BDA0003762336660000074
When one is a double bond, the other is
Figure BDA0003762336660000075
Is a single bond and R 6 Is absent; and when two are
Figure BDA0003762336660000076
When all are single bonds, then R 6 Is hydrogen.
In some embodiments, R 1 Is an alkyl group. In some embodiments, R 1 Is an unsubstituted alkyl group. In some embodiments, R 1 Is C 1 -C 6 An alkyl group. In some embodiments, R 1 is-CH 3 、–CF 3 or-CH 2 CH 3 . In some embodiments, R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 Alkyl (e.g., -CH) 3 )。
In some embodiments, R 2 Is alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl and R 3 Is hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl or R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered ring.
In some embodiments, R 2 Is hydrogen or alkyl. In some embodiments, R 2 And R 3 Each independently hydrogen or alkyl. In some embodiments, R 2 And R 3 Each independently is hydrogen or C 1 -C 6 A haloalkyl group. In some embodiments, R 2 And R 3 Each independently is hydrogen, -CF 3 or-CH 3 . In some embodiments, R 4 is-OH or halogen. In some embodiments, R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group. In some embodiments, R 4 Is hydrogen and R 5 Is a halogen. In some embodiments, R 4 And R 5 Is halogen. In some embodiments, R 4 And R 5 Is hydrogen. In some embodiments, R 2 Is aryl or heteroaryl and R 3 Is hydrogen. In some embodiments, R 2 Is carbocyclyl or heterocyclyl and R 3 Is hydrogen. In some embodiments, R 2 And R 3 Is hydrogen. In some embodiments, R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered carbocyclic or heterocyclic ring.
In some embodiments, the compound of formula (I-67) is selected from compounds of formula (I-A67), (I-B67), or (I-C67):
Figure BDA0003762336660000081
in some embodiments, the compound of formula (I-67) is selected from compounds of formula (I-A67):
Figure BDA0003762336660000082
in some embodiments, the compound of formula (I-67) is selected from compounds of formula (I-C67):
Figure BDA0003762336660000083
in one aspect, provided herein is a compound selected from the group consisting of:
Figure BDA0003762336660000084
Figure BDA0003762336660000091
Figure BDA0003762336660000101
Figure BDA0003762336660000111
Figure BDA0003762336660000121
Figure BDA0003762336660000131
Figure BDA0003762336660000141
Figure BDA0003762336660000151
Figure BDA0003762336660000161
Figure BDA0003762336660000171
Figure BDA0003762336660000181
in one aspect, provided herein is a pharmaceutical composition comprising a compound described herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In one aspect, provided herein is a method of inducing sedation or anesthesia comprising administering to a subject an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
In one aspect, provided herein is a method of treating or preventing a disease described herein, comprising administering to a subject in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
In some embodiments, the disease is a metabolic disease.
In some embodiments, the disease is an autoimmune disease.
In some embodiments, the disease is rheumatoid arthritis, juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis, crohn's disease, ulcerative colitis, and plaque psoriasis.
In some embodiments, the disease is a Gastrointestinal (GI) disorder such as constipation, irritable Bowel Syndrome (IBS), inflammatory Bowel Disease (IBD) (e.g., ulcerative colitis, crohn's disease), a structural disorder affecting the gastrointestinal tract, an anal disorder (e.g., hemorrhoids, internal hemorrhoids, external hemorrhoids, anal fissure, perianal abscess, anal fistula), colonic polyps, cancer, or colitis.
In some embodiments, the disease is inflammatory bowel disease.
In some embodiments, the disease is cancer, diabetes, or a sterol synthesis disorder.
In one aspect, provided herein is a method of treating or preventing a CNS-related disorder, comprising administering to a subject in need thereof an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. In some embodiments, the CNS-related disorders are adjustment disorders, anxiety disorders (including obsessive-compulsive disorders, post-traumatic stress disorder, and social phobia), cognitive disorders (including alzheimer's disease and other forms of dementia (e.g., frontotemporal dementia), separation disorders, eating disorders, mood disorders (including depression (e.g., postpartum depression), bipolar disorders, dysthymic disorders, suicidal tendencies), schizophrenia or other psychotic disorders (including schizoaffective disorders), sleep disorders (including insomnia), substance-related disorders, personality disorders (including obsessive-compulsive personality disorders), autism spectrum disorders (including those associated with mutations in Shank family proteins (e.g., shank 3)), neurodevelopmental disorders (including Rett syndrome, complex sarcoidosis), multiple sclerosis, sterol synthesis disorders, pain (including acute and chronic pain; headache, e.g., migraine), encephalopathy secondary to medical disorders (including hepatic encephalopathy and anti-NMDA receptor encephalitis), acute liver failure, glycine encephalopathy, epilepsy (including status epilepticus and monogenic forms of epilepsy, such as Dravet disease), stroke, traumatic brain injury, movement disorders (including huntington's disease and parkinson's disease), vision disorders, hearing loss, or tinnitus.
In some embodiments, the disease is huntington's disease. In some embodiments, the disease is parkinson's disease. In some embodiments, the disease is an inflammatory disease (e.g., lupus).
In some embodiments, the disease is a sterol synthesis disorder.
In some embodiments, the disease is Smith-Lemli-optitz syndrome (SLOS). In some embodiments, the disease is streptosterol disease. In some embodiments, the disease is sitosterolemia. In some embodiments, the disease is brain tendon xanthomatosis (CTX). In some embodiments, the disease is Mevalonate Kinase Deficiency (MKD). In some embodiments, the disease is SC4MOL gene mutation (SMO Deficiency). In some embodiments, the disease is Niemann-Pick disease. In some embodiments, the disease is Autism Spectrum Disorder (ASD). In some embodiments, the disease is associated with phenylketonuria.
In one aspect, provided herein is a method of effecting negative allosteric modulation of an NMDA receptor in a subject comprising administering to the subject a compound described herein, e.g., a compound of formula (a), a compound of formula (I-63), or a compound of formula (I-67).
Definition of
Chemical definition
The definitions of specific functional groups and chemical terms are described in more detail below. Chemical elements are defined according to the CAS version of the periodic table of elements on page 75 of the Handbook of Chemistry and philics, and specific functional groups are also defined as described herein in general. Furthermore, in Thomas Sorrell, organic Chemistry, university Science Books, sausaltito, 1999; smith and March, march's Advanced Organic Chemistry, 5 th edition, john Wiley&Sons,Inc.,New York,2001;Larock,Comprehensive Organic Transformations,VCHpublishers,Inc.,New York,1989; and Carruther, some Modern Methods of Organic Synthesis,3 rd The general principles of organic chemistry, as well as specific functional groups and reactivity, are described in Edition, cambridge university, cambridge, 1987.
Isomers may be separated from mixtures by methods known to those skilled in the art, including chiral High Pressure Liquid Chromatography (HPLC) and the formation and crystallization of chiral salts; alternatively, the preferred isomers may be prepared by asymmetric synthesis. See, for example: jacques et al, enantiomers, racemates and solutions (Wiley Interscience, new York, 1981); wilen et al, tetrahedron 33:2725 (1977); eliel, stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and Wilen, tablets of solving Agents and optical solutions, page 268 (E.L. Eliel, ed., univ.of Notre Damepress, notre Dame, IN 1972). The invention also includes compounds described herein as individual isomers that are substantially free of other isomers or include compounds that are mixtures of different isomers.
As used herein, "enantiomeric excess" ("e.e.") or "% enantiomeric excess" ("% e.e.") of a composition refers to one enantiomer in excess relative to the other enantiomer present in the composition. For example, the composition may contain 90% of one enantiomer, e.g., the S enantiomer, and 10% of the other enantiomer, i.e., the R enantiomer.
e.e.=(90-10)/100=80%。
Thus, a composition containing 90% of one enantiomer and 10% of the other enantiomer is said to have an enantiomeric excess of 80%.
As used herein, a "diastereomeric excess" ("d.e.") or "% diastereomeric excess" ("% d.e.") of a composition refers to an excess of one diastereomer relative to one or more different diastereomers present in the composition. For example, a composition may contain 90% of one diastereomer and 10% of one or more different diastereomer.
d.e.=(90-10)/100=80%。
Thus, a composition containing 90% of one diastereomer and 10% of one or more different diastereomer is said to have a diastereomeric excess of 80%.
The absolute configuration of the asymmetric center can be determined using methods known to those skilled in the art. In some embodiments, the absolute configuration of the asymmetric center in the compound can be elucidated from the X-ray single crystal structure of the compound. In some embodiments, the absolute configuration of an asymmetric center as illustrated by the X-ray crystal structure of a compound can be used to infer the absolute configuration of the corresponding asymmetric center in another compound obtained from the same or similar synthetic method. In some embodiments, the absolute configuration of the asymmetric center as illustrated by the X-ray crystal structure of the compound can be used in conjunction with spectroscopic techniques (e.g., NMR spectroscopy, e.g., 1 H NMR spectroscopy or 19 F NMR spectroscopy) in combination with inferring the absolute configuration of the corresponding asymmetric center in the other compound.
When a range of values is recited, it is intended to include all values and subranges within the range. For example, "C 1-6 Alkyl "is intended to cover C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 1-6 、C 1-5 、C 1-4 、C 1-3 、C 1-2 、C 2-6 、C 2-5 、C 2-4 、C 2-3 、C 3-6 、C 3-5 、C 3-4 、C 4-6 、C 4-5 And C 5-6 An alkyl group.
The following terms are intended to have the meanings indicated below, and to aid in understanding the description of the invention and the intended scope. When describing the present invention, which can include compounds, pharmaceutical compositions comprising the compounds, and methods of using the compounds and compositions, the following terms (if any) have the following meanings, unless otherwise indicated. It is also to be understood that any of the moieties defined below may be substituted with a variety of substituents when described herein, and each definition is intended to include such substituted moieties within its scope as defined below. Unless otherwise indicated, the term "substituted" is defined as follows. It is further understood that the terms "group" and "radical" as used herein may be considered interchangeable. The articles "a" and "an" may be used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. For example, "an analog" refers to one or more than one analog.
"aliphatic group" means an alkyl, alkenyl, alkynyl, or carbocyclyl group as defined herein.
"alkyl" means a straight or branched chain saturated hydrocarbon radical having from 1 to 20 carbon atoms ("C) 1-20 Alkyl "). In some embodiments, the alkyl group has 1 to 12 carbon atoms ("C) 1-12 Alkyl "). In some embodiments, the alkyl group has 1 to 10 carbon atoms ("C) 1-10 Alkyl "). In some embodiments, the alkyl group has 1 to 9 carbon atoms ("C) 1-9 Alkyl "). In some embodiments, the alkyl group has 1 to 8 carbon atoms ("C) 1-8 Alkyl "). In some embodiments, the alkyl group has 1 to 7 carbon atoms ("C) 1-7 Alkyl "). In some embodiments, the alkyl group has 1 to 6 carbon atoms ("C) 1-6 Alkyl ", also referred to herein as" lower alkyl "). In some embodiments, the alkyl group has 1 to 5 carbon atoms ("C) 1-5 Alkyl "). In some embodiments, the alkyl group has 1 to 4 carbon atoms ("C) 1-4 Alkyl "). In some embodiments, the alkyl group has 1 to 3 carbon atoms ("C) 1-3 Alkyl "). In some embodiments, the alkyl group has 1 to 2 carbon atoms ("C) 1-2 Alkyl "). In some embodiments, the alkyl group has 1 carbon atom ("C) 1 Alkyl "). In some embodiments, the alkyl group has 2 to 6 carbon atoms ("C) 2-6 Alkyl "). C 1-6 Examples of alkyl groups include methyl (C) 1 ) Ethyl (C) 2 ) N-propyl (C) 3 ) Isopropyl (C) 3 ) N-butyl (C) 4 ) Tert-butyl (C) 4 ) Sec-butyl (C) 4 ) Isobutyl (C) 4 ) N-pentyl group (C) 5 ) 3-pentyl radical (C) 5 ) Pentyl group (C) 5 ) Isopentyl group (C) 5 ) Neopentyl (C) 5 ) 3-methyl-2-butyl (C) 5 ) Tert-amyl (C) 5 ) And n-hexyl (C) 6 ). Other examples of alkyl groups include n-heptyl (C) 7 ) N-octyl (C) 8 ) And the like. Unless otherwise specified, an alkyl group is independently in each instance optionally substituted, i.e., unsubstituted (an "unsubstituted alkyl") or substituted (a "substituted alkyl") with one or more substituents (e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent). In some embodiments, the alkyl group is unsubstituted C 1-10 Alkyl (e.g. -CH) 3 ). In some embodiments, the alkyl group is substituted C 1-10 An alkyl group. Common alkyl abbreviations include Me (-CH) 3 )、Et(-CH 2 CH 3 )、iPr(-CH(CH 3 ) 2 )、nPr(-CH 2 CH 2 CH 3 )、n-Bu(-CH 2 CH 2 CH 2 CH 3 ) Or i-Bu (-CH) 2 CH(CH 3 ) 2 )。
"alkylene" refers to an alkyl group in which two hydrogens are removed to provide a divalent group, and which may be substituted or unsubstituted. Unsubstituted alkylene groups include, but are not limited to, methylene (-CH) 2 -) ethylene (-CH 2 CH 2 -), propylene (-CH) 2 CH 2 CH 2 -), butylene (-CH) 2 CH 2 CH 2 CH 2 -) pentylene (-CH) 2 CH 2 CH 2 CH 2 CH 2 -), hexylene (-CH) 2 CH 2 CH 2 CH 2 CH 2 CH 2 -) and the like. Exemplary substituted alkylene groups, such as substituted with one or more alkyl (methyl) groups, include, but are not limited to, substituted methylene (-CH (CH) 3 )-、(-C(CH 3 ) 2 -) substituted ethylene (-CH (CH) 3 )CH 2 -、-CH 2 CH(CH 3 )-、-C(CH 3 ) 2 CH 2 -、-CH 2 C(CH 3 ) 2 -) substituted propylene (-CH (CH) 3 )CH 2 CH 2 -、-CH 2 CH(CH 3 )CH 2 -、-CH 2 CH 2 CH(CH 3 )-、-C(CH 3 ) 2 CH 2 CH 2 -、-CH 2 C(CH 3 ) 2 CH 2 -、-CH 2 CH 2 C(CH 3 ) 2 -) and the like. When a range or value of carbon is given for a particular alkylene group, it is understood that the range or value refers to the range or value of carbon on a straight carbon divalent chain. The alkylene groups can be substituted with one or more substituents described herein or can be unsubstituted.
"alkenyl" means a group of straight or branched hydrocarbon radicals having from 2 to 20 carbon atoms, one or more carbon-carbon double bonds (e.g., 1, 2, 3, or 4 carbon-carbon double bonds), and optionally one or more carbon-carbon triple bonds (e.g., 1, 2, 3, or 4 carbon-carbon triple bonds) ("C) 2-20 Alkenyl "). In some embodiments, the alkenyl group does not comprise any triple bonds. In some embodiments, an alkenyl group has 2 to 10 carbon atoms ("C) 2-10 Alkenyl "). In some embodiments, an alkenyl group has 2 to 9 carbon atoms ("C) 2-9 Alkenyl "). In some embodiments, an alkenyl group has 2 to 8 carbon atoms ("C) 2-8 Alkenyl "). In some embodiments, an alkenyl group has 2 to 7 carbon atoms ("C) 2-7 Alkenyl "). In some embodiments, an alkenyl group has 2 to 6 carbon atoms ("C) 2-6 Alkenyl "). In some embodiments, an alkenyl group has 2 to 5 carbon atoms ("C) 2-5 Alkenyl "). In some embodiments, an alkenyl group has 2 to 4 carbon atoms ("C) 2-4 Alkenyl "). In some embodiments, an alkenyl group has 2 to 3 carbon atoms ("C) 2-3 Alkenyl "). In some embodiments, an alkenyl group has 2 carbon atoms ("C) 2 Alkenyl "). The one or more carbon-carbon double bonds may be internal (as in 2-butenyl) or terminal (as in 1-butenyl). C 2-4 Examples of alkenyl groups include vinyl (C) 2 ) 1-propenyl group (C) 3 ) 2-propenyl (C) 3 ) 1-butenyl (C) 4 ) 2-butenyl (C) 4 ) Butadienyl radical (C) 4 ) And the like. C 2-6 Examples of alkenyl groups include the foregoing C 2-4 Alkenyl radical and pentenyl radical (C) 5 ) Pentadienyl radical (C) 5 ) Hexenyl (C) 6 ) And so on. Other examples of alkenyl include heptenyl (C) 7 ) Octenyl (C) 8 ) Octrienyl (C) 8 ) And so on. Unless otherwise specified, each alkenyl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted alkenyl") or substituted (a "substituted alkenyl") with one or more substituents (e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent). In some embodiments, the alkenyl group is unsubstituted C 2-10 An alkenyl group. In some embodiments, the alkenyl group is substituted C 2-10 An alkenyl group.
"alkynyl" refers to a group having a straight or branched hydrocarbon radical ("C") of 2 to 20 carbon atoms, one or more carbon-carbon triple bonds (e.g., 1, 2, 3, or 4 carbon-carbon triple bonds), and optionally one or more carbon-carbon double bonds (e.g., 1, 2, 3, or 4 carbon-carbon double bonds) 2-20 Alkynyl "). In some embodiments, alkynyl groups do not contain any double bonds. In some embodiments, alkynyl groups have 2 to 10 carbon atoms ("C) 2-10 Alkynyl "). In some embodiments, alkynyl groups have 2 to 9 carbon atoms ("C) 2-9 Alkynyl "). In some embodiments, alkynyl groups have 2 to 8 carbon atoms ("C) 2-8 Alkynyl "). In some embodiments, alkynyl groups have 2 to 7 carbon atoms ("C) 2-7 Alkynyl "). In some embodiments, alkynyl groups have 2 to 6 carbon atoms ("C) 2-6 Alkynyl "). In some embodiments, alkynyl groups have 2 to 5 carbon atoms ("C) 2-5 Alkynyl "). In some embodiments, alkynyl groups have 2 to 4 carbon atoms ("C) 2-4 Alkynyl "). In some embodiments, alkynyl groups have 2 to 3 carbon atoms ("C) 2-3 Alkynyl "). In some embodiments, alkynyl groups have 2 carbon atoms ("C) 2 Alkynyl "). The one or more carbon-carbon triple bonds may be internal (as in 2-butynyl) or terminal (as in 1-butynyl). C 2-4 Examples of alkynyl groups include, but are not limited to: ethynyl (C) 2 ) 1-propynyl (C) 3 ) 2-propynyl (C) 3 ) 1-butynyl (C) 4 ) 2-butynyl (C) 4 ) And the like. C 2-6 Examples of alkynyl groups include the foregoing C 2-4 Alkynyl radical and pentynyl (C) 5 ) Hexynyl (C) 6 ) And the like. Other examples of alkynyl groups include heptynyl (C) 7 ) Octynyl (C) 8 ) And so on. Unless otherwise specified, each alkynyl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted alkynyl") or substituted (a "substituted alkynyl") with one or more substituents, e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent. In some embodiments, the alkynyl group is unsubstituted C 2-10 Alkynyl. In some embodiments, the alkynyl group is substituted C 2-10 Alkynyl.
The term "heteroalkyl", as used herein, refers to an alkyl group, as defined herein, which further comprises one or more (e.g., 1, 2, 3, or 4) heteroatoms (e.g., oxygen, sulfur, nitrogen, boron, silicon, phosphorus) in the parent chain, wherein the one or more heteroatoms are interposed between adjacent carbon atoms in the parent carbon chain, and/or one or more heteroatoms are interposed between a carbon atom and the parent molecule, i.e., between the points of attachment. In some embodiments, a heteroalkyl group refers to a saturated group having 1 to 10 carbon atoms and 1, 2, 3, or 4 heteroatoms ("heteroc 1-10 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 1 to 9 carbon atoms and 1, 2, 3, or 4 heteroatoms ("heteroc) 1-9 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 1 to 8 carbon atoms and 1, 2, 3, or 4 heteroatoms ("heteroc) 1-8 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 1 to 7 carbon atoms and 1, 2, 3, or 4 heteroatoms ("heteroc) 1-7 Alkyl "). In some embodiments, a heteroalkyl group is a group having 1 to 6 carbon atoms and 1, 2, or 3 heteroatoms ("heteroc) 1-6 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 1 to 5 carbon atoms and 1 or 2 heteroatoms ("heteroc) 1-5 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 1 to 4 carbon atoms and 1 or 2 heteroatoms ("heteroc) 1-4 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 1 to 3 carbon atoms and 1 heteroatom ("heteroc) 1-3 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 1 to 2 carbon atoms and 1 heteroatom ("heteroc) 1-2 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 1 carbon atom and 1 heteroatom ("heteroc 1 Alkyl "). In some embodiments, a heteroalkyl group is a saturated group having 2 to 6 carbon atoms and 1 or 2 heteroatoms ("heteroc) 2-6 Alkyl "). Unless otherwise specified, heteroalkyl groups are each independently unsubstituted (an "unsubstituted heteroalkyl") or substituted (a "substituted heteroalkyl") with one or more substituents. In some embodiments, the heteroalkyl group is unsubstituted heteroc 1-10 An alkyl group. In some embodiments, the heteroalkyl group is a substituted heteroc 1-10 An alkyl group.
"aryl" refers to a monocyclic or polycyclic (e.g., bicyclic or tricyclic) group of an 4n +2 aromatic ring system (e.g., having 6, 10, or 14 pi electrons shared in a ring arrangement) and having 6 to 14 ring carbon atoms and 0 heteroatom in the aromatic ring system ("C) 6-14 Aryl "). In some embodiments, an aryl group has six ring carbon atoms ("C) 6 Aryl "; such as phenyl). In some embodiments, an aryl group has ten ring carbon atoms ("C) 10 Aryl "; such as naphthyl groups such as 1-naphthyl and 2-naphthyl). In some embodiments, an aryl group has fourteen ring carbon atoms ("C) 14 Aryl "; such as an anthracene group). "aryl" also includes ring systems in which an aryl ring, as defined above, is fused to one or more carbocyclic or heterocyclic groups in which the linking group or point is on the aryl ring, and in which case the number of carbon atoms continues to be specified as the number of carbon atoms in the aryl ring system. Typical aryl groups include, but are not limited to, groups derived from: aceanthrylene (acenaphthylene), acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, naphthalene, and naphthalene,
Figure BDA0003762336660000252
(chrysene), coronene (coronene), fluoranthene, fluorene, hexacene (hexacene), hexaphene (hexaphene), hexalene (hexalene), asymmetric indacene (as-indacene), symmetric indacene (s-indacene), indane, indene, naphthalene, octocane (octacene), octacene (octacene), octotam (octalene), octacene (octalene), penta-2, 4-diene, pentacene (pentalene), dicyclopentadiene (pentalene), dibenzophenanthrene (pentalene), perylene (phenalene), phenalene (phenalene), phenanthrene, picene (picene), heptalene (pyradene), pyrene, anthracene (anthrene), rubicene (triphenylene), triphenylene (triphenylene), and triphenylene (triphenylene). Specific aryl groups include phenyl, naphthyl, indenyl, and tetrahydronaphthyl. Unless otherwise specified, each aryl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted aryl") or substituted (a "substituted aryl") with one or more substituents. In some embodiments, the aryl group is unsubstituted C 6-14 And (3) an aryl group. In some embodiments, the aryl group is substituted C 6-14 And (4) an aryl group.
In some embodiments, the aryl group is substituted with one or more substituents selected from halogen, C 1 -C 8 Alkyl radical, C 1 -C 8 Haloalkyl, cyano, hydroxy, C 1 -C 8 Alkoxy and amino groups.
Representative examples of substituted aryl groups include the following:
Figure BDA0003762336660000251
wherein R is 56 And R 57 One of which may be hydrogen and R 56 And R 57 At least one is independently selected from C 1 -C 8 Alkyl radical, C 1 -C 8 Haloalkyl, 4-to 10-membered heterocyclyl, alkanoyl, C 1 -C 8 Alkoxy, heteroaryloxy, alkylamino, arylamino, heteroarylamino, NR 58 COR 59 、NR 58 SOR 59 、NR 58 SO 2 R 59 COOalkyl, COOaryl, CONR 58 R 59 、CONR 58 OR 59 、NR 58 R 59 、SO 2 NR 58 R 59 S-alkyl, SO 2 Alkyl, S aryl, SO 2 An aryl group; or R 56 And R 57 Can be combined to form a cyclic ring of 5 to 8 atoms (saturated or unsaturated), optionally containing one or more heteroatoms selected from N, O or S. R 60 And R 61 Independently of one another is hydrogen, C 1 -C 8 Alkyl radical, C 1 -C 4 Haloalkyl, C 3 -C 10 Cycloalkyl, 4-to 10-membered heterocyclyl, C 6 -C 10 Aryl, substituted C 6 -C 10 Aryl, 5-to 10-membered heteroaryl, or substituted 5-to 10-membered heteroaryl.
"fused aryl" refers to an aryl group, wherein two of the ring carbon atoms are shared with a second aryl or heteroaryl ring or with a carbocyclyl or heterocyclyl ring.
"heteroaryl" refers to a group having a 5 to 10 membered monocyclic or bicyclic 4n +2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic arrangement) with ring carbon atoms and 1 to 4 ring heteroatoms in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5 to 10 membered heteroaryl"). In heteroaryl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as valency permits. Heteroaryl bicyclic ring systems can contain one or more heteroatoms within one or both rings. "heteroaryl" includes ring systems in which a heteroaryl ring, as defined above, is fused to one or more carbocyclyl or heterocyclyl groups, wherein the point of attachment is on the heteroaryl ring, and in which case the number of ring members continues to be specified as the number of ring members in the heteroaryl ring system. "heteroaryl" also includes ring systems in which a heteroaryl ring, as defined above, is fused to one or more aryl groups, wherein the point of attachment is on the aryl or heteroaryl ring, and in which case the number of ring members is specified as the number of ring members in the fused (aryl/heteroaryl) ring system. Bicyclic heteroaryl groups (e.g., indolyl, quinolinyl, carbazolyl, etc.) wherein one ring does not contain a heteroatom, the point of attachment may be on one of the two rings, i.e., on the ring with the heteroatom (e.g., 2-indolyl) or on the ring without the heteroatom (e.g., 5-indolyl).
In some embodiments, the heteroaryl group is a 5 to 10 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5 to 10 membered heteroaryl"). In some embodiments, the heteroaryl group is a 5 to 8 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms within the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5 to 8 membered heteroaryl"). In some embodiments, the heteroaryl group is a 5 to 6 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5 to 6 membered heteroaryl"). In some embodiments, the 5-to 6-membered heteroaryl has 1-3 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-to 6-membered heteroaryl has 1-2 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-to 6-membered heteroaryl has 1 ring heteroatom selected from nitrogen, oxygen, and sulfur. Unless otherwise specified, each heteroaryl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted heteroaryl") or substituted (a "substituted heteroaryl") with one or more substituents. In some embodiments, the heteroaryl group is an unsubstituted 5 to 14 membered heteroaryl. In some embodiments, the heteroaryl group is a substituted 5-to 14-membered heteroaryl.
Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to, pyrrolyl, furanyl, and thienyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to, triazolyl, oxadiazolyl, and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to, tetrazolyl. Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to, pyridinyl. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to, pyridazinyl, pyrimidinyl, and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively. Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to, azepinyl, oxacycloheptatrienyl, and thiacycloheptatrienyl. Exemplary 5, 6-bicyclic heteroaryl groups include, but are not limited to, indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl, benzoisothiofuranyl, benzimidazolyl, benzoxazolyl, benzoisoxazolyl, benzooxadiazolyl, benzothiazolyl, benzoisothiazolyl, benzothiadiazolyl, indolizinyl, and purinyl. Exemplary 6,6-bicyclic heteroaryl groups include, but are not limited to, naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl, and quinazolinyl.
Examples of representative heteroaryl groups include the following:
Figure BDA0003762336660000271
wherein each Z is selected from the group consisting of carbonyl, N, NR 65 O and S; and R is 65 Independently of one another is hydrogen, C 1 -C 8 Alkyl radical, C 3 -C 10 Cycloalkyl, 4-to 10-membered heterocyclic group, C 6 -C 10 Aryl and 5 to 10 membered heteroaryl.
"carbocyclyl" or "carbocycle" refers to a non-aromatic ring system having from 3 to 10 ring carbon atoms ("C) 3-10 Carbocyclyl ") and 0 heteroatom of a non-aromatic cyclic hydrocarbon group. In some embodiments, carbocyclyl groups have 3 to 8 ring carbon atoms ("C) 3-8 Carbocyclyl "). In some embodiments, carbocyclyl groups have 3 to 7 ring carbon atoms ("C) 3-7 Carbocyclyl "). In some embodiments, carbocyclyl groups have 3 to 6 ring carbon atoms ("C) 3-6 Carbocyclyl "). In some embodiments, carbocyclyl groups have 5 to 10 ring carbon atoms ("C) 5-10 Carbocyclic group"). Exemplary C 3-6 Carbocyclyl groups include, but are not limited to, cyclopropyl (C) 3 ) Cyclopropenyl group (C) 3 ) Cyclobutyl (C) 4 ) Cyclobutenyl radical (C) 4 ) Cyclopentyl (C) 5 ) Cyclopentenyl group (C) 5 ) Cyclohexyl (C) 6 ) Cyclohexenyl (C) 6 ) Cyclohexadienyl (C) 6 ) And the like. Exemplary C 3-8 Carbocyclyl groups include, but are not limited to, C as previously described 3-6 Carbocyclyl group and cycloheptyl (C) 7 ) Cycloheptenyl (C) 7 ) Cycloheptadienyl (C) 7 ) Cycloheptatrienyl (C) 7 ) Cyclooctyl (C) 8 ) Cyclooctenyl (C) 8 ) Bicyclo [2.2.1 ] s]Heptyl (C) 7 ) Bicyclo [2.2.2]Octyl radical (C) 8 ) And the like. Exemplary C 3-10 Carbocyclyl groups include, but are not limited to, C as described above 3-8 Carbocyclyl group and cyclononyl (C) 9 ) Cyclononenyl (C) 9 ) Cyclodecyl (C) 10 ) Cyclodecenyl (C) 10 ) octahydro-1H-indenyl (C) 9 ) Decahydronaphthyl (C) 10 ) Spiro [4.5 ]]Decyl (C) 10 ) And the like. As described in the previous examples, in some embodiments, carbocyclyl groups are monocyclic ("monocyclic carbocyclyl") or contain a fused, bridged, or spiro ring system (e.g., a bicyclic system ("bicyclic carbocyclyl")) and may be saturated or may be partially unsaturated. "carbocyclyl" also includes ring systems in which a carbocyclic ring, as described above, is fused to one or more aryl or heteroaryl groups, wherein the point of attachment is on the carbocyclic ring, and in which case the number of carbons continues to be specified as the number of carbons in the carbocyclic ring system. Unless otherwise specified, each carbocyclyl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted carbocyclyl") or substituted with one or more substituents (a "substituted carbocyclyl"). In some embodiments, carbocyclyl groups are unsubstituted C 3-10 A carbocyclic group. In some embodiments, carbocyclyl group is substituted C 3-10 A carbocyclic group.
In some embodiments, "carbocyclyl" is a monocyclic saturated carbocyclyl group having from 3 to 10 ring carbon atoms ("C) 3-10 Cycloalkyl "). In some embodiments, a cycloalkyl groupThe radicals having from 3 to 8 ring carbon atoms ("C) 3-8 Cycloalkyl "). In some embodiments, cycloalkyl groups have 3 to 6 ring carbon atoms ("C) 3-6 Cycloalkyl "). In some embodiments, cycloalkyl groups have 5 to 6 ring carbon atoms ("C) 5-6 Cycloalkyl "). In some embodiments, cycloalkyl groups have from 5 to 10 ring carbon atoms ("C) 5-10 Cycloalkyl "). C 5-6 Examples of cycloalkyl radicals include cyclopentyl (C) 5 ) And cyclohexyl (C) 6 )。C 3-6 Examples of cycloalkyl radicals include the aforementioned C 5-6 Cycloalkyl radical and cyclopropyl (C) 3 ) And cyclobutyl (C) 4 )。C 3-8 Examples of cycloalkyl radicals include the aforementioned C 3-6 Cycloalkyl radical and cycloheptyl (C) 7 ) And cyclooctyl (C) 8 ). Unless otherwise specified, each cycloalkyl group is independently unsubstituted (an "unsubstituted cycloalkyl") or substituted (a "substituted cycloalkyl") with one or more substituents. In some embodiments, the cycloalkyl group is unsubstituted C 3-10 A cycloalkyl group. In some embodiments, the cycloalkyl group is substituted C 3-10 A cycloalkyl group.
"heterocyclyl" or "heterocyclic" refers to a group of 3 to 10 membered non-aromatic ring systems having ring carbon atoms and 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon ("3 to 10 membered heterocyclyl"). In heterocyclyl groups containing one or more nitrogen atoms, the point of attachment may be a carbon or nitrogen atom, as valency permits. A heterocyclyl group may be monocyclic ("monocyclic heterocyclyl") or a fused, bridged or spiro ring system (e.g., a bicyclic system ("bicyclic heterocyclyl")) and may be saturated or may be partially unsaturated. The heterocyclyl bicyclic ring system may contain one or more heteroatoms in one or both rings. "heterocyclyl" also includes ring systems in which a heterocyclyl ring as defined above is fused to one or more carbocyclyl groups, where the point of attachment is on the carbocyclyl or on the heterocyclyl ring, or includes ring systems in which a heterocyclyl ring as defined above is fused to one or more aryl or heteroaryl groups, where the point of attachment is on the heterocyclyl ring, and in that case, the number of ring members continues to be specified as the number of ring members in the heterocyclyl ring system. Unless otherwise specified, each instance of a heterocyclyl is independently optionally substituted, i.e., unsubstituted (an "unsubstituted heterocyclyl") or substituted (a "substituted heterocyclyl") with one or more substituents. In some embodiments, a heterocyclyl group is an unsubstituted 3-to 10-membered heterocyclyl. In some embodiments, the heterocyclyl group is a substituted 3-to 10-membered heterocyclyl.
In some embodiments, a heterocyclyl group is a 5-to 10-membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon ("5-to 10-membered heterocyclyl"). In some embodiments, a heterocyclyl group is a 5-to 8-membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-to 8-membered heterocyclyl"). In some embodiments, a heterocyclyl group is a 5-to 6-membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-to 6-membered heterocyclyl"). In some embodiments, the 5-to 6-membered heterocyclyl has 1-3 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-to 6-membered heterocyclyl has 1-2 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-to 6-membered heterocyclyl has one ring heteroatom selected from nitrogen, oxygen, and sulfur.
Exemplary 3-membered heterocyclyl groups containing one heteroatom include, but are not limited to, aziridinyl, oxacyclopropaneyl, thiacyclopropaneyl. Exemplary 4-membered heterocyclyl groups containing one heteroatom include, but are not limited to, azetidinyl, oxetanyl and thietanyl. Exemplary 5-membered heterocyclyl groups containing one heteroatom include, but are not limited to, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl, and pyrrole-2, 5-dione. Exemplary 5-membered heterocyclyl groups containing two heteroatoms include, but are not limited to, dioxolanyl, oxathiolanyl (oxathiacyclopentyl), dithiolanyl (disulphenyl) And oxazolidin-2-ones. Exemplary 5-membered heterocyclyl groups containing three heteroatoms include, but are not limited to, triazolinyl, oxadiazolinyl, and thiadiazolinyl. Exemplary 6-membered heterocyclyl groups containing one heteroatom include, but are not limited to, piperidinyl, tetrahydropyranyl, dihydropyridinyl, and thiacyclohexyl. Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, but are not limited to, piperazinyl, morpholinyl, dithiacyclohexyl, dioxanyl. Exemplary 6-membered heterocyclyl groups containing three heteroatoms include, but are not limited to, triazacyclohexyl (triazonyl). Exemplary 7-membered heterocyclyl groups containing one heteroatom include, but are not limited to, azepanyl, oxepinyl, and thiepanyl. Exemplary 8-membered heterocyclyl groups containing one heteroatom include, but are not limited to, azacyclooctyl, oxocyclooctyl, and thiepinyl. Exemplary with C 6 Aryl ring fused 5-membered heterocyclyl groups (also referred to herein as 5, 6-bicyclic heterocycles) include, but are not limited to, indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinonyl (benzoxazolinonyl), and the like. Exemplary 6-membered heterocyclyl groups fused to an aryl ring (also referred to herein as 6,6-bicyclic heterocycles) include, but are not limited to, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
"Nitrogen-containing heterocyclyl" refers to 4-to 7-membered non-aromatic cyclic groups containing at least one nitrogen atom, such as, but not limited to, morpholine, piperidine (e.g., 2-piperidinyl, 3-piperidinyl, and 4-piperidinyl), pyrrolidine (e.g., 2-pyrrolidinyl and 3-pyrrolidinyl), azetidine, pyrrolidone, imidazoline, imidazolidinone, 2-pyrazoline, pyrazolidine, piperazine, and N-alkylpiperazine such as N-methylpiperazine. Specific examples include azetidines, piperidones, and piperazinones.
When used to describe a compound or group present on a compound, "hetero" means that one or more carbon atoms in the compound or group are replaced with a nitrogen, oxygen, or sulfur heteroatom. Hetero can be used to describe any of the hydrocarbon groups described above, such as alkyl (e.g., heteroalkyl), cycloalkyl (e.g., heterocyclyl), aryl (e.g., heteroaryl), cycloalkenyl (e.g., cycloheteroalkenyl), and the like, having from 1 to 5, specifically from 1 to 3 heteroatoms.
"acyl" refers to-C (O) R 20 Group (I) wherein R 20 Is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, as defined herein. "alkanoyl" is an acyl group wherein R 20 Are groups other than hydrogen. Representative acyl groups include, but are not limited to, formyl (-CHO), acetyl (-C (= O) CH 3 ) Cyclohexylcarbonyl group, cyclohexylmethylcarbonyl group, benzoylgroup (-C (= O) Ph), benzylcarbonyl group (-C (= O) CH 2 Ph)、--C(O)-C 1 -C 8 Alkyl, -C (O) - (CH) 2 ) t (C 6 -C 10 Aryl), -C (O) - (CH) 2 ) t (5-to 10-membered heteroaryl), -C (O) - (CH) 2 ) t (C 3 -C 10 Cycloalkyl) and-C (O) - (CH) 2 ) t (4-to 10-membered heterocyclyl), wherein t is an integer of 0 to 4. In some embodiments, R 21 Is C substituted by halogen or hydroxy 1 -C 8 An alkyl group; or C 3 -C 10 Cycloalkyl, 4-to 10-membered heterocyclyl, C 6 -C 10 Aryl, arylalkyl, 5-to 10-membered heteroaryl or heteroarylalkyl, each of which is unsubstituted C 1 -C 4 Alkyl, halogen, unsubstituted C 1 -C 4 Alkoxy, unsubstituted C 1 -C 4 Haloalkyl, unsubstituted C 1 -C 4 Hydroxyalkyl or unsubstituted C 1 -C 4 Haloalkoxy or hydroxy.
"alkoxy" means a group-OR 29 Wherein R is 29 Is a substituted or unsubstituted alkyl group, a substituted or unsubstituted alkenyl group, a substituted or unsubstituted alkynyl group, a substituted or unsubstituted carbocyclyl group, a substituted or unsubstituted heterocyclyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted heteroaryl group. Specific alkoxy groups are methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy and 1, 2-dimethylbutoxy. In particular Alkoxy groups are lower alkoxy, i.e. having 1 to 6 carbon atoms. Further specific alkoxy groups have 1 to 4 carbon atoms.
In some embodiments, R 29 Is a group having one or more substituents, for example 1 to 5 substituents, and specifically 1 to 3 substituents, especially 1 substituent, selected from amino, substituted amino, C 6 -C 10 Aryl, aryloxy, carboxy, cyano, C 3 -C 10 Cycloalkyl, 4-to 10-membered heterocyclyl, halogen, 5-to 10-membered heteroaryl, hydroxy, nitro, thioalkoxy, thioaryloxy, mercapto, alkyl-S (O) -, aryl-S (O) -, alkyl-S (O) 2 And aryl-S (O) 2 -. Exemplary 'substituted alkoxy' groups include, but are not limited to, -O- (CH) 2 ) t (C 6 -C 10 Aryl), -O- (CH) 2 ) t (5-to 10-membered heteroaryl), -O- (CH) 2 ) t (C 3 -C 10 Cycloalkyl) and-O- (CH) 2 ) t (4-to 10-membered heterocyclyl), wherein t is an integer from 0 to 4, and any aryl, heteroaryl, cycloalkyl or heterocyclyl groups present may themselves be unsubstituted C 1 -C 4 Alkyl, halogen, unsubstituted C 1 -C 4 Alkoxy, unsubstituted C 1 -C 4 Haloalkyl, unsubstituted C 1 -C 4 Hydroxyalkyl or unsubstituted C 1 -C 4 Haloalkoxy or hydroxy. A specific exemplary 'substituted alkoxy' is-OCF 3 、-OCH 2 CF 3 、-OCH 2 Ph、-OCH 2 -cyclopropyl, -OCH 2 CH 2 OH and-OCH 2 CH 2 NMe 2
"amino" refers to the group-NH 2
"oxo" means-C (= O) -.
"substituted amino" refers to the formula-N (R) 38 ) 2 Wherein R is 38 Is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclylOr an unsubstituted aryl, substituted or unsubstituted heteroaryl, or amino protecting group, wherein at least one R 38 Is not hydrogen. In some embodiments, each R is independently selected from R, and R 38 Independently selected from hydrogen, C 1 -C 8 Alkyl radical, C 3 -C 8 Alkenyl radical, C 3 -C 8 Alkynyl, C 6 -C 10 Aryl, 5-to 10-membered heteroaryl, 4-to 10-membered heterocyclyl or C 3 -C 10 A cycloalkyl group; or C 1 -C 8 Alkyl substituted with halogen or hydroxy; c 3 -C 8 Alkenyl substituted with halogen or hydroxy; c 3 -C 8 Alkynyl substituted with halogen or hydroxy; or- (CH) 2 ) t (C 6 -C 10 Aryl), - (CH) 2 ) t (5-to 10-membered heteroaryl), - (CH) 2 ) t (C 3 -C 10 Cycloalkyl) or- (CH) 2 ) t (4-to 10-membered heterocyclyl), wherein t is an integer from 0 to 8, each of which is unsubstituted C 1 -C 4 Alkyl, halogen, unsubstituted C 1 -C 4 Alkoxy, unsubstituted C 1 -C 4 Haloalkyl, unsubstituted C 1 -C 4 Hydroxyalkyl or unsubstituted C 1 -C 4 Haloalkoxy or hydroxy substitution; or two R 38 The groups combine to form an alkylene group.
Exemplary "substituted amino" includes, but is not limited to, -NR 39 -C 1 -C 8 Alkyl, -NR 39 -(CH 2 ) t (C 6 -C 10 Aryl), -NR 39 -(CH 2 ) t (5-to 10-membered heteroaryl), -NR 39 -(CH 2 ) t (C 3 -C 10 Cycloalkyl) and-NR 39 -(CH 2 ) t (4-to 10-membered heterocyclyl), where t is an integer from 0 to 4, e.g. 1 or 2, each R 39 Independently represent H or C 1 -C 8 An alkyl group; and any alkyl groups present may themselves be substituted by halogen, substituted or unsubstituted amino or hydroxy; and any aryl, heteroaryl, cycloalkyl or heterocyclyl groups present may themselves be unsubstituted C 1 -C 4 Alkyl, halogenUnsubstituted C 1 -C 4 Alkoxy, unsubstituted C 1 -C 4 Haloalkyl, unsubstituted C 1 -C 4 Hydroxyalkyl or unsubstituted C 1 -C 4 Haloalkoxy or hydroxy. For the avoidance of doubt, the term 'substituted amino' includes: alkylamino, substituted alkylamino, alkylarylamino, substituted alkylarylamino, arylamino, substituted arylamino, dialkylamino and substituted dialkylamino groups as defined below. Substituted amino groups include mono-substituted amino and di-substituted amino groups.
"carboxy" refers to a-C (O) OH group.
"cyano" refers to the group-CN.
"halo" or "halogen" refers to fluoro (F), chloro (Cl), bromo (Br), and iodo (I). In some embodiments, the halo group is fluoro or chloro.
"haloalkyl" refers to an alkyl group wherein the alkyl group is substituted with one or more halogens. Typical haloalkyl groups include, but are not limited to, trifluoromethyl (-CF) 3 ) Difluoromethyl (-CHF) 2 ) Fluoromethyl (-CH) 2 F) Chloromethyl (-CH) 2 Cl), dichloromethyl (-CHCl) 2 ) Tribromomethyl (-CH) 2 Br), etc.
"hydroxy" refers to the group-OH.
"Nitro" means the radical-NO 2
"Thioketo (Thioketo)" means a group = S.
"carbocyclylalkyl" refers to an alkyl group in which the alkyl group is substituted with a cycloalkyl group. Common carbocyclylalkyl groups include, but are not limited to, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, cycloheptylmethyl, cyclooctylmethyl, cyclopropylethyl, cyclobutylethyl, cyclopentylethyl, cyclohexylethyl, cycloheptylethyl, and cyclooctylethyl and the like.
"Heterocyclylalkyl" refers to an alkyl group in which the alkyl group is substituted with a heterocyclyl group. Common heterocyclylalkyl groups include, but are not limited to, pyrrolidinylmethyl, piperidinylmethyl, piperazinylmethyl, morpholinylmethyl, pyrrolidinylethyl, piperidinylethyl, piperazinylethyl, morpholinylethyl, and the like.
"aralkyl" is a subset of alkyl and aryl groups defined herein, and refers to an optionally substituted alkyl group substituted with an optionally substituted aryl group.
Alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl groups, as defined herein, are optionally substituted (e.g., "substituted" or "unsubstituted" alkyl, "substituted" or "unsubstituted" alkenyl, "substituted" or "unsubstituted" alkynyl, "substituted" or "unsubstituted" carbocyclyl, "substituted" or "unsubstituted" heterocyclyl, "substituted" or "unsubstituted" aryl, or "substituted" or "unsubstituted" heteroaryl groups). Generally, the term "substituted", whether preceded by the term "optionally" or not, means that at least one hydrogen present on a group (e.g., a carbon or nitrogen atom) is replaced with an allowable substituent, e.g., a substituent that replaces a substituent that results in a stable compound (e.g., a compound that does not spontaneously undergo transformation, e.g., by rearrangement, cyclization, elimination or other reaction). Unless otherwise specified, a "substituted" group has a substituent at one or more substitutable positions of the group, and when more than one position in any given structure is substituted, the substituents at each position are the same or different. The term "substituted" is considered to include substitution by all permissible substituents of organic compounds, any of which described herein, resulting in the formation of stable compounds. The present invention contemplates any and all such combinations to obtain stable compounds. For purposes herein, a heteroatom such as nitrogen may have a hydrogen substituent and/or any suitable substituent as described herein that satisfies the valency of the heteroatom and results in the formation of a stable moiety.
Exemplary carbon atom substituents include, but are not limited to: halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OR aa 、-ON(R bb ) 2 、-N(R bb ) 2 、-N(R bb ) 3 + X - 、-N(OR cc )R bb 、-SH、-SR aa 、-SSR cc 、-C(=O)R aa 、-CO 2 H、-CHO、-C(OR cc ) 2 、-CO 2 R aa 、-OC(=O)R aa 、-OCO 2 R aa 、-C(=O)N(R bb ) 2 、-OC(=O)N(R bb ) 2 、-NR bb C(=O)R aa 、-NR bb CO 2 R aa 、-NR bb C(=O)N(R bb ) 2 、-C(=NR bb )R aa 、-C(=NR bb )OR aa 、-OC(=NR bb )R aa 、-OC(=NR bb )OR aa 、-C(=NR bb )N(R bb ) 2 、-OC(=NR bb )N(R bb ) 2 、-NR bb C(=NR bb )N(R bb ) 2 、-C(=O)NR bb SO 2 R aa 、-NR bb SO 2 R aa 、-SO 2 N(R bb ) 2 、-SO 2 R aa 、-SO 2 OR aa 、-OSO 2 R aa 、-S(=O)R aa 、-OS(=O)R aa 、-Si(R aa ) 3 、-OSi(R aa ) 3 -C(=S)N(R bb ) 2 、-C(=O)SR aa 、-C(=S)SR aa 、-SC(=S)SR aa 、-SC(=O)SR aa 、-OC(=O)SR aa 、-SC(=O)OR aa 、-SC(=O)R aa 、-P(=O) 2 R aa 、-OP(=O) 2 R aa 、-P(=O)(R aa ) 2 、-OP(=O)(R aa ) 2 、-OP(=O)(OR cc ) 2 、-P(=O) 2 N(R bb ) 2 、-OP(=O) 2 N(R bb ) 2 、-P(=O)(NR bb ) 2 、-OP(=O)(NR bb ) 2 、-NR bb P(=O)(OR cc ) 2 、-NR bb P(=O)(NR bb ) 2 、-P(R cc ) 2 、-P(R cc ) 3 、-OP(R cc ) 2 、-OP(R cc ) 3 、-B(R aa ) 2 、-B(OR cc ) 2 、-BR aa (OR cc )、C 1-10 Alkyl radical, C 1-10 Haloalkyl, C 2-10 Alkenyl radical, C 2-10 Alkynyl, C 3-10 Carbocyclyl, 3-to 14-membered heterocyclyl, C 6-14 Aryl and 5 to 14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1, 2, 3, 4 or 5R dd Substituted by groups;
or two geminal hydrogens on a carbon atom are replaced by a group = O, = S, = NN (R) bb ) 2 、=NNR bb C(=O)R aa 、=NNR bb C(=O)OR aa 、=NNR bb S(=O) 2 R aa 、=NR bb Or = NOR cc Replacing;
R aa independently at each occurrence is selected from C 1-10 Alkyl radical, C 1-10 Haloalkyl, C 2-10 Alkenyl radical, C 2-10 Alkynyl, C 3-10 Carbocyclyl, 3-to 14-membered heterocyclyl, C 6-14 Aryl and 5-to 14-membered heteroaryl, or two R aa Combine to form a 3-to 14-membered heterocyclyl or 5-to 14-membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5R dd Substituted by groups;
R bb independently at each occurrence, is selected from hydrogen, -OH, -OR aa 、-N(R cc ) 2 、-CN、-C(=O)R aa 、-C(=O)N(R cc ) 2 、-CO 2 R aa 、-SO 2 R aa 、-C(=NR cc )OR aa 、-C(=NR cc )N(R cc ) 2 、-SO 2 N(R cc ) 2 、-SO 2 R cc 、-SO 2 OR cc 、-SOR aa 、-C(=S)N(R cc ) 2 、-C(=O)SR cc 、-C(=S)SR cc 、-P(=O) 2 R aa 、-P(=O)(R aa ) 2 、-P(=O) 2 N(R cc ) 2 、-P(=O)(NR cc ) 2 、C 1-10 Alkyl radical, C 1-10 Haloalkyl group、C 2-10 Alkenyl radical, C 2-10 Alkynyl, C 3-10 Carbocyclyl, 3-to 14-membered heterocyclyl, C 6-14 Aryl and 5-to 14-membered heteroaryl, or two R bb The groups combine to form a 3-to 14-membered heterocyclyl or 5-to 14-membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1, 2, 3, 4 or 5R dd Substitution of radicals;
R cc independently at each occurrence, selected from hydrogen, C 1-10 Alkyl radical, C 1-10 Haloalkyl, C 2-10 Alkenyl radical, C 2-10 Alkynyl, C 3-10 Carbocyclyl, 3-to 14-membered heterocyclyl, C 6-14 Aryl and 5-to 14-membered heteroaryl, or two R cc The groups combine to form a 3-to 14-membered heterocyclyl or 5-to 14-membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1, 2, 3, 4 or 5R dd Substituted by groups;
R dd independently at each occurrence, is selected from halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OR ee 、-ON(R ff ) 2 、-N(R ff ) 2 、-N(R ff ) 3 + X-、-N(OR ee )R ff 、-SH、-SR ee 、-SSR ee 、-C(=O)R ee 、-CO 2 H、-CO 2 R ee 、-OC(=O)R ee 、-OCO 2 R ee 、-C(=O)N(R ff ) 2 、-OC(=O)N(R ff ) 2 、-NR ff C(=O)R ee 、-NR ff CO 2 R ee 、-NR ff C(=O)N(R ff ) 2 、-C(=NR ff )OR ee 、-OC(=NR ff )R ee 、-OC(=NR ff )OR ee 、-C(=NR ff )N(R ff ) 2 、-OC(=NR ff )N(R ff ) 2 、-NR ff C(=NR ff )N(R ff ) 2 ,-NR ff SO 2 R ee 、-SO 2 N(R ff ) 2 、-SO 2 R ee 、-SO 2 OR ee 、-OSO 2 R ee 、-S(=O)R ee 、-Si(R ee ) 3 、-OSi(R ee ) 3 、-C(=S)N(R ff ) 2 、-C(=O)SR ee 、-C(=S)SR ee 、-SC(=S)SR ee 、-P(=O) 2 R ee 、-P(=O)(R ee ) 2 、-OP(=O)(R ee ) 2 、-OP(=O)(OR ee ) 2 、C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-10 Carbocyclyl, 3-to 10-membered heterocyclyl, C 6-10 Aryl, 5 to 10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1, 2, 3, 4 or 5R gg Substituted by radicals, or two geminal R dd Substituents may combine to form = O or = S;
R ee independently at each occurrence is selected from C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-10 Carbocyclyl, C 6-10 Aryl, 3-to 10-membered heterocyclyl, and 3-to 10-membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5R gg Substitution of radicals;
R ff Independently at each occurrence, selected from hydrogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-10 Carbocyclyl, 3-to 10-membered heterocyclyl, C 6-10 Aryl and 5 to 10 membered heteroaryl, or two R ff The groups combine to form a 3-to 14-membered heterocyclyl or 5-to 14-membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1, 2, 3, 4 or 5R gg Substituted by groups; and is
R gg Independently at each occurrence is halogen, -CN, -NO 2 、-N 3 、-SO 2 H、-SO 3 H、-OH、-OC 1-6 Alkyl, -ON (C) 1-6 Alkyl radical) 2 、-N(C 1-6 Alkyl radical) 2 、-N(C 1-6 Alkyl radical) 3 + X-、-NH(C 1-6 Alkyl radical) 2 + X-、-NH 2 (C 1-6 Alkyl radical) + X - 、-NH 3 + X - 、-N(OC 1-6 Alkyl) (C) 1-6 Alkyl), -N (OH) (C) 1-6 Alkyl), -NH (OH), -SH, -SC 1-6 Alkyl, -SS (C) 1-6 Alkyl), -C (= O) (C) 1-6 Alkyl), -CO 2 H、-CO 2 (C 1-6 Alkyl), -OC (= O) (C) 1-6 Alkyl), -OCO 2 (C 1-6 Alkyl), -C (= O) NH 2 、-C(=O)N(C 1-6 Alkyl radical) 2 、-OC(=O)NH(C 1-6 Alkyl), -NHC (= O) (C) 1-6 Alkyl), -N (C) 1-6 Alkyl) C (= O) (C 1-6 Alkyl), -NHCO 2 (C 1-6 Alkyl), -NHC (= O) N (C) 1-6 Alkyl radical) 2 、-NHC(=O)NH(C 1-6 Alkyl), -NHC (= O) NH 2 、-C(=NH)O(C 1-6 Alkyl), -OC (= NH) (C) 1-6 Alkyl), -OC (= NH) OC 1-6 Alkyl, -C (= NH) N (C) 1-6 Alkyl radical) 2 、-C(=NH)NH(C 1-6 Alkyl), -C (= NH) NH 2 、-OC(=NH)N(C 1-6 Alkyl radical) 2 、-OC(NH)NH(C 1-6 Alkyl), -OC (NH) NH 2 、-NHC(NH)N(C 1-6 Alkyl radical) 2 、-NHC(=NH)NH 2 、-NHSO 2 (C 1-6 Alkyl), -SO 2 N(C 1-6 Alkyl radical) 2 、-SO 2 NH(C 1-6 Alkyl), -SO 2 NH 2 ,-SO 2 C 1-6 Alkyl, -SO 2 OC 1-6 Alkyl, -OSO 2 C 1-6 Alkyl, -SOC 1-6 Alkyl, -Si (C) 1-6 Alkyl radical) 3 、-OSi(C 1-6 Alkyl radical) 3 -C(=S)N(C 1-6 Alkyl radical) 2 、C(=S)NH(C 1-6 Alkyl), C (= S) NH 2 、-C(=O)S(C 1-6 Alkyl), -C (= S) SC 1-6 Alkyl group, -SC (= S) SC 1-6 Alkyl, -P (= O) 2 (C 1-6 Alkyl), -P (= O) (C) 1-6 Alkyl radical) 2 、-OP(=O)(C 1-6 Alkyl radical) 2 、-OP(=O)(OC 1-6 Alkyl radical) 2 、C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-10 Carbocyclyl, C 6-10 Aryl, 3-to 10-membered heterocyclyl, 5-to 10-membered heteroaryl; or two twinned R gg Substituents may combine to form = O or = S; wherein X - Are counter ions.
"counterions" or "anionic counterions" are negatively charged groups that associate with the cationic quaternary amino groups to maintain electrical neutrality. Exemplary counterions include halide ions (e.g., F) - 、Cl - 、Br - 、I - )、NO 3 - 、ClO 4 - 、OH - 、H 2 PO 4 - 、HSO 4 - 、SO 4 -2 Sulfonate ions (e.g., methanesulfonate, trifluoromethanesulfonate, p-toluenesulfonate, benzenesulfonate, 10-camphorsulfonate, naphthalene-2-sulfonate, naphthalene-1-sulfonic acid-5-sulfonate, ethane-1-sulfonic acid-2-sulfonate, etc.) and carboxylate ions (e.g., acetate, propionate, benzoate, glycerate, lactate, tartrate, glycolate, etc.).
The nitrogen atoms may be substituted or unsubstituted as valency permits, and include primary, secondary, tertiary, and quaternary nitrogen atoms. Exemplary nitrogen atom substituents include, but are not limited to, hydrogen, -OH, -OR aa 、-N(R cc ) 2 、-CN、-C(=O)R aa 、-C(=O)N(R cc ) 2 、-CO 2 R aa 、-SO 2 R aa 、-C(=NR bb )R aa 、-C(=NR cc )OR aa 、-C(=NR cc )N(R cc ) 2 、-SO 2 N(R cc ) 2 、-SO 2 R cc 、-SO 2 OR cc 、-SOR aa 、-C(=S)N(R cc ) 2 、-C(=O)SR cc 、-C(=S)SR cc 、-P(=O) 2 R aa 、-P(=O)(R aa ) 2 、-P(=O) 2 N(R cc ) 2 、-P(=O)(NR cc ) 2 、C 1-10 Alkyl radical, C 1-10 Haloalkyl, C 2-10 Alkenyl radical, C 2-10 Alkynyl, C 3-10 Carbocyclyl, 3-to 14-membered heterocyclyl, C 6-14 Aryl and 5-to 14-membered heteroaryl, or two R's bound to a nitrogen atom cc The groups combine to form a 3-to 14-membered heterocyclyl or 5-to 14-membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1, 2, 3, 4 or 5R dd Is substituted by radicals in which R aa 、R bb 、R cc And R dd As described above.
These and other exemplary substituents are described in more detail in the detailed description, examples and claims. The present invention is not intended to be limited in any way to the exemplary substituents listed above.
Other definitions
The term "pharmaceutically acceptable salts" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are known in the art. For example, pharmaceutically acceptable salts are described in detail by Berge et al in J.pharmaceutical Sciences (1977) 66. Pharmaceutically acceptable salts of the compounds of the present invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, non-toxic acid addition salts are salts of amino groups with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods in the art such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, citrates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, formates Fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, embonate, pectate (pectinate), persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Pharmaceutically acceptable salts derived from suitable bases include alkali metal salts, alkaline earth metal salts, ammonium salts and N + (C 1-4 Alkyl radical) 4 And (3) salt. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium and the like. Pharmaceutically acceptable salts also include, when appropriate, the non-toxic ammonium, quaternary ammonium and amine cation salts formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
A "subject" intended for administration includes, but is not limited to, a human (i.e., a male or female of any age group, such as a pediatric subject (e.g., an infant, a child, a juvenile) or an adult subject (e.g., an adolescent, a middle-aged, or an elderly human)) and/or a non-human animal, such as a mammal (e.g., a primate (e.g., a cynomolgus monkey, a rhesus monkey), a cow, a pig, a horse, a sheep, a goat, a rodent, a cat, and/or a dog). In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal. The terms "human," "patient," and "subject" are used interchangeably herein.
Diseases, disorders, and conditions are used interchangeably herein.
As used herein and unless otherwise specified, the terms "treat," "treating," and "treatment" refer to an action taken when a subject suffers from a particular disease, disorder, or condition that reduces the severity of the disease, disorder, or condition, or delays or retards the progression of the disease, disorder, or condition ("therapeutic treatment"), as well as an action before the subject begins to suffer from the particular disease, disorder, or condition ("prophylactic treatment").
Generally, an "effective amount" of a compound is an amount sufficient to elicit a desired biological response. One skilled in the art will appreciate that the effective amount of a compound of the present invention may vary depending on the following factors: the desired biological endpoint, the pharmacokinetics of the compound, the disease being treated, the mode of administration, and the age, health, and condition of the subject. An effective amount encompasses both therapeutic and prophylactic treatment.
As used herein and unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic effect or to delay or minimize one or more symptoms associated with a disease, disorder or condition in the treatment of the disease, disorder or condition. A therapeutically effective amount of a compound refers to an amount that provides a therapeutic effect in the treatment of a disease, disorder, or condition, when the therapeutic agent is used alone or in combination with other therapies. The term "therapeutically effective amount" can include an amount that generally improves the treatment, reduces or avoids the symptoms or causes of a disease or disorder, or increases the efficacy of another therapeutic agent.
As used herein and unless otherwise specified, a "prophylactically effective amount" of a compound is an amount sufficient to prevent or avoid recurrence of a disease, disorder, or condition or one or more symptoms associated with the disease, disorder, or condition. A prophylactically effective amount of a compound is an amount that provides a prophylactic effect in the prevention of a disease, disorder, or condition when the therapeutic agent is used alone or in combination with other agents. The term "prophylactically effective amount" can include an amount that generally improves prophylaxis or increases the prophylactic effect of another prophylactic agent.
Detailed description of some embodiments of the invention
As generally described herein, the present invention provides substituted oxysterol for use in the prevention and/or treatment of various diseases, including but not limited to NMDA-mediated diseases. These compounds are expected to exhibit improved in vivo efficacy, pharmacokinetic (PK) profile, oral bioavailability, formulation, stability and/or safety compared to other oxysterol.
Compound (I)
In one aspect, provided herein is a compound according to formula (a):
Figure BDA0003762336660000381
or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen or alkyl (e.g. C) 1 -C 6 Alkyl); r 2 And R 3 Each independently hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl or heteroaryl, or R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring; r 4 And R 5 Each independently is hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl), or R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group; r 6 Absent or hydrogen; r is G Is hydrogen or alkyl; and is provided with
Figure BDA0003762336660000391
Represents a single or double bond, wherein
Figure BDA0003762336660000392
When one is a double bond, the other is
Figure BDA0003762336660000393
Is a single bond and R 6 Is absent; and when two are
Figure BDA0003762336660000394
When all are single bonds, then R 6 Is hydrogen.
In some embodiments, R 1 Is alkyl (e.g. C) 1 -C 6 Alkyl). In some embodiments, R 1 Is C 1 -C 6 Alkyl (e.g., -CH) 3 、–CH 2 CH 3 、–CH 2 OCH 3 or-CF 3 ). In some embodiments, R 1 is-CH 3 、–CF 3 or-CH 2 CH 3 . In some embodiments, R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl groups).
In some embodiments, R 2 Is hydrogen or alkyl (e.g. C) 1 -C 6 Alkyl groups).
In some embodiments, R 2 And R 3 Each independently hydrogen or alkyl (e.g., C) 1 -C 6 Alkyl groups). In some embodiments, R 2 And R 3 Each independently is hydrogen or C 1 -C 6 Haloalkyl (e.g., -CF) 3 ). In some embodiments, R 2 And R 3 Each independently is hydrogen, -CF 3 or-CH 3
In some embodiments, R 4 is-OH or halogen (e.g., -F).
In some embodiments, R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group.
In some embodiments, R 4 Is hydrogen and R 5 Is halogen (e.g., -F). In some embodiments, R 4 And R 5 Is halogen (e.g., -F). In some embodiments, R 4 And R 5 Is hydrogen.
In some embodiments, R 2 Is aryl or heteroaryl and R 3 Is hydrogen. In some embodiments, R 2 Is carbocyclyl or heterocyclyl and R 3 Is hydrogen. In some embodiments, R 2 And R 3 Is hydrogen. In some embodiments, R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered carbocyclic or heterocyclic ring.
In some embodiments, R 6 Is hydrogen and
Figure BDA0003762336660000395
represents a single bond.
In some embodiments, R G Is hydrogen or-CH 3
In one aspect, provided herein are compounds according to formula (I-63):
Figure BDA0003762336660000396
or a pharmaceutically acceptable salt thereof, wherein: r is 1 Is alkyl (e.g. C) 1 -C 6 Alkyl groups); r is 2 And R 3 Each independently hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), alkenyl (e.g. C) 2 -C 6 Alkenyl), alkynyl (e.g., C) 2 -C 6 Alkynyl), carbocyclyl, heterocyclyl, aryl or heteroaryl, or R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring; r 4 And R 5 Each independently hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl), or R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group; r is 6 Absent or hydrogen; and is provided with
Figure BDA0003762336660000401
Represents a single or double bond, wherein
Figure BDA0003762336660000402
When one is a double bond, the other is
Figure BDA0003762336660000403
Is a single bond and R 6 Is absent; and when two are
Figure BDA0003762336660000404
When all are single bonds, then R 6 Is hydrogen.
In some embodiments, R 1 Is alkyl (e.g. C) 1 -C 6 Alkyl). In some embodiments, R 1 Is C 1 -C 6 Alkyl (e.g., -CH) 3 、–CH 2 CH 3 、–CH 2 OCH 3 or-CF 3 ). In some embodiments, R 1 is-CH 3 、–CF 3 or-CH 2 CH 3 . In some embodiments, R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl groups). In some embodiments, R 1 Is an unsubstituted alkyl group. In some embodiments R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 Alkyl (e.g., -CH) 3 )。
In some embodiments, R 2 Is alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl and R 3 Is hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl or heteroaryl or R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered ring.
In some embodiments, R 2 Is alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl and R 3 Is hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl or R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered ring.
In some embodiments, R 2 Is hydrogen or alkyl (e.g. C) 1 -C 6 Alkyl groups).
In some embodiments, R 2 And R 3 Each independently hydrogen or alkyl (e.g., C) 1 -C 6 Alkyl groups). In some embodiments, R 2 And R 3 Each independently is hydrogen or C 1 -C 6 Haloalkyl (e.g., -CF) 3 ). In some embodiments, R 2 And R 3 Each independently is C 5 Alkyl (e.g., substituted or unsubstituted isoamyl) or hydrogen. In some embodiments, R 2 And R 3 Each independently is an isoamyl group (e.g., a substituted or unsubstituted isoamyl group) or hydrogen. In some embodiments, R 2 And R 3 Each independently is hydrogen, -CF 3 or-CH 3
In some embodiments, R 4 is-OH or halogen (e.g., -F).
In some embodiments, R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group. In some embodiments, R 4 Is hydrogen and R 5 Is halogen (e.g., -F).
In some embodiments, R 4 And R 5 Is halogen (e.g., -F). In some embodiments, R 4 And R 5 Is hydrogen.
In some embodiments, R 2 Is aryl or heteroaryl and R 3 Is hydrogen. In some embodiments, R 2 Is carbocyclyl or heterocyclyl and R 3 Is hydrogen. In some embodiments, R 2 And R 3 Is hydrogen. In some embodiments, R 2 Is an isopentyl group (e.g., a substituted or unsubstituted isopentyl group) and R 3 Is hydrogen. In some embodiments, R 2 is-CF 3 or-CH 3 And R is 3 Is hydrogen or-CH 3 . In some embodiments, R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered carbocyclic or heterocyclic ring.
In some embodiments, R 1 is-CH 3 or-CH 2 CH 3 ,R 2 Is an isoamyl group (e.g., a substituted or unsubstituted isoamyl group), and R 3 Is hydrogen. In some embodiments, R 1 is-CH 3 or-CH 2 CH 3 ,R 2 Is unsubstituted isopentyl, and R 3 Is hydrogen.
In some embodiments, R 2 Is unsubstituted C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group. In thatIn some embodiments, R 2 Is unsubstituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is a pyridyl group. In some embodiments, each R is 2 Is isoamyl and R 3 Is hydrogen. In some embodiments, R 2 is-CF 3 And R is 3 Is hydrogen. In some embodiments, R 2 Is unsubstituted alkyl (e.g., unsubstituted C) 1 -C 6 Alkyl groups). In some embodiments, R 2 Is carbocyclylalkyl. In some embodiments, R 2 Is carbocyclylalkyl and R 3 Is hydrogen. In some embodiments, R 2 Is an aralkyl group (e.g., benzyl). In some embodiments, R 2 Is a heterocyclylalkyl group. In some embodiments, wherein R is 2 Is unsubstituted C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, carbocyclyl, carbocyclylalkyl, aralkyl, or heterocyclylalkyl.
In some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-A63), (I-B63), or (I-C63):
Figure BDA0003762336660000411
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-A63):
Figure BDA0003762336660000412
In some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-B63):
Figure BDA0003762336660000421
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-C63):
Figure BDA0003762336660000422
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-D63):
Figure BDA0003762336660000423
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-E63):
Figure BDA0003762336660000424
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-D-I63) or (I-D-ii 63):
Figure BDA0003762336660000425
in some embodiments, the compound of formula (I-63) is selected from compounds of formula (I-E-I63) or (I-E-ii 63):
Figure BDA0003762336660000431
in one aspect, provided herein are compounds according to formula (I-67):
Figure BDA0003762336660000432
or a pharmaceutically acceptable salt thereof, wherein: r 1 Is hydrogen or alkyl (e.g. C) 1 -C 6 Alkyl groups); r is 2 And R 3 Each independently hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl, or R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring; r 4 And R 5 Each independently hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl (e.g. C) 1 -C 3 Alkyl), or R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group; r is 6 Absent or hydrogen; and is provided with
Figure BDA0003762336660000433
Represents a single or double bond, wherein
Figure BDA0003762336660000434
When one is a double bond, the other is
Figure BDA0003762336660000435
Is a single bond and R 6 Is absent; and when two are
Figure BDA0003762336660000436
When all are single bonds, then R 6 Is hydrogen.
In some embodiments, R 1 Is an alkyl group. In some embodiments, R 1 Is an unsubstituted alkyl group. In some embodiments, R 1 Is C 1 -C 6 An alkyl group. In some embodiments, R 1 is-CH 3 、–CF 3 or-CH 2 CH 3 . In some embodiments, R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 Alkyl (e.g., -CH) 3 )。
In some embodiments, R 2 Is alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl and R 3 Is hydrogen, alkyl (e.g. C) 1 -C 6 Alkyl), carbocyclyl, heterocyclyl, aryl or heteroaryl or R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered ring.
In some embodiments, R 2 Is hydrogen or alkyl. In some embodiments, R 2 And R 3 Each independently hydrogen or alkyl. In a 1In some embodiments, R 2 And R 3 Each independently is hydrogen or C 1 -C 6 A haloalkyl group. In some embodiments, R 2 And R 3 Each independently is hydrogen, -CF 3 or-CH 3 . In some embodiments, R 4 is-OH or halogen. In some embodiments, R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group. In some embodiments, R 4 Is hydrogen and R 5 Is halogen. In some embodiments, R 4 And R 5 Is a halogen. In some embodiments, R 4 And R 5 Is hydrogen. In some embodiments, R 2 Is aryl or heteroaryl and R 3 Is hydrogen. In some embodiments, R 2 Is carbocyclyl or heterocyclyl and R 3 Is hydrogen. In some embodiments, R 2 And R 3 Is hydrogen. In some embodiments, R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered carbocyclic or heterocyclic ring.
In some embodiments, the compound of formula (I-67) is selected from compounds of formula (I-A67), (I-B67), or (I-C67):
Figure BDA0003762336660000441
in some embodiments, the compound of formula (I-67) is selected from compounds of formula (I-A67):
Figure BDA0003762336660000442
in some embodiments, the compound of formula (I-67) is selected from compounds of formula (I-C67):
Figure BDA0003762336660000443
exemplary compounds of the invention include:
Figure BDA0003762336660000451
Figure BDA0003762336660000461
Figure BDA0003762336660000471
Figure BDA0003762336660000481
Figure BDA0003762336660000491
Figure BDA0003762336660000501
Figure BDA0003762336660000511
Figure BDA0003762336660000521
Figure BDA0003762336660000531
Figure BDA0003762336660000541
alternative embodiments
In an alternative embodiment, the compounds described herein may also comprise one or more isotopic substitutions. For example, hydrogen may be 2 H (D or deuterium) or 3 H (T or tritium); the carbon may be, for example, 13 c or 14 C; the oxygen may be, for example, 18 o; the nitrogen may be, for example, 15 n, etc. In other embodiments, the specific isotope (e.g., 3 H, 13 C, 14 C, 18 o, or 15 N) may represent at least 1%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or at least 99.9% of the total isotopic abundance of the element occupying a particular site of the compound.
Pharmaceutical composition
In another aspect, the invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof.
When used as a medicament, the compounds provided herein are typically administered in the form of a pharmaceutical composition. The composition may be prepared in a manner well known in the pharmaceutical art and comprises at least one active compound.
In one embodiment, with respect to the pharmaceutical composition, the carrier is a parenteral carrier, oral or topical carrier.
The invention also relates to a compound as described herein or a pharmaceutical composition thereof for use as a medicament or drug.
Typically, the compounds provided herein are administered in a therapeutically effective amount. The amount of compound actually administered will generally be determined by a physician, in the light of the relevant circumstances, including the condition being treated, the chosen route of administration, the compound actually administered, the age, weight and response of the individual patient, the severity of the patient's symptoms, and the like.
The pharmaceutical compositions provided herein can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. Depending on the desired route of delivery, the compounds provided herein are preferably formulated as injectable or oral compositions, or as saline, lotions or patches for transdermal administration.
Compositions for oral administration may take the form of bulk liquid solutions or suspensions or bulk powders. More commonly, however, the composition is presented in unit dosage form to aid in accurate administration. The term "unit dosage form" refers to a single dose of physically discrete units suitable for use in human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of liquid compositions, or pills, tablets, or capsules and the like in the case of solid compositions. In such compositions, the compound is typically a minor component (about 0.1 to 50% or preferably about 1 to about 40% by weight), the remainder being various vehicles or carriers, and processing aids that aid in forming the desired dosage form.
Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous vehicles, as well as buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like. Solid forms may include, for example, any of the following ingredients or compounds having similar properties: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; excipients, such as starch or lactose; disintegrating agents, such as alginic acid, primogel or corn starch; lubricants, such as magnesium stearate; glidants, such as colloidal silicon dioxide; sweetening agents, such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate or orange flavoring.
Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline or other injectable carriers known in the art. As before, the active compound in the composition is typically a minority component, typically about 0.05 to 10% by weight, the remainder being injectable carriers and the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient in an amount generally from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight. When formulated as an ointment, the active ingredient is typically combined with a paraffinic or water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream, together with, for example, an oil-in-water cream base. Such transdermal formulations are known in the art and typically contain other ingredients to increase the transdermal penetration stability of the active ingredient or formulation. All such known transdermal formulations and ingredients are included within the scope of the present disclosure.
The compounds provided herein can also be administered via a transdermal device. Thus, transdermal administration can be accomplished by using a patch of the depot or porous membrane type or by using a solid matrix species.
The above-described ingredients for orally-administrable, injectable, or topically-administrable compositions are merely representative. Other materials and processing techniques are listed in section 8 of Remington' pharmaceutical Sciences,17 th edition, 1985, mackpublishing company, easton, pennsylvania, which is incorporated herein by reference, among others.
The above ingredients for orally administrable, injectable or topically administrable compositions are merely representative. Other materials and processing techniques are listed in section 8 of Remington's The Science and Practice of Pharmacy,21 st edition, 2005, publisher: lippincott Williams & Wilkins, which is incorporated herein by reference, among others.
The compounds of the present invention may be administered in sustained release form, or by sustained release drug delivery systems. A description of representative sustained release materials can be found in Remington' pharmaceutical Sciences.
The invention also relates to pharmaceutically acceptable formulations of the compounds described herein. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins, which consist of 6, 7 and 8 α -l, 4-linked glucose units, respectively, optionally containing one or more substituents on the linked sugar moiety including, but not limited to, methylated, hydroxyalkylated, acylated and sulfoalkyl ether substituents. In some embodiments, the cyclodextrin is a sulfoalkyl ether Beta-cyclodextrins, such as, for example, sulfobutylether beta-cyclodextrin, also known as
Figure BDA0003762336660000571
See, e.g., U.S.5,376,645. In some embodiments, the formulation comprises hexapropyl- β -cyclodextrin. In a more specific embodiment, the formulation comprises hexapropyl- β -cyclodextrin (10-50% aqueous solution).
The invention also relates to pharmaceutically acceptable acid addition salts of the compounds described herein. Acids that can be used to prepare pharmaceutically acceptable salts are those that form non-toxic acid addition salts, i.e., salts containing pharmaceutically acceptable anions, such as hydrochloride, hydroiodide, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, maleate, fumarate, benzoate, p-toluenesulfonate, and the like.
The following formulation examples illustrate representative pharmaceutical compositions that may be prepared according to the present invention. However, the present invention is not limited to the following pharmaceutical compositions.
Exemplary formulation 1-tablet: the compounds described herein, or pharmaceutically acceptable salts thereof, can be mixed as a dry powder with a dry gelatin binder in a weight ratio of about 1. A small amount of magnesium stearate may be added as a lubricant. The mixture is formed into tablets of 240 to 270mg (each having 80 to 90mg of active compound) in a tablet press.
Exemplary formulation 2-capsule: the compounds described herein, or pharmaceutically acceptable salts thereof, can be mixed as a dry powder with a starch diluent in a weight ratio of about 1. The mixture was filled into 250mg capsules (125 mg of active compound in each capsule).
Exemplary formulation 3-liquid: the compound described herein or a pharmaceutically acceptable salt thereof (125 mg) may be mixed with sucrose (1.75 g) and xanthan gum (4 mg), and the resulting mixture may be blended, sieved through a 10 mesh US sieve, and then mixed with an aqueous solution of microcrystalline cellulose and sodium carboxymethylcellulose (11, 89, 50 mg) prepared previously. Sodium benzoate (10 mg), flavouring and colouring agents were diluted with water and added with stirring. Sufficient water may then be added to produce a total volume of 5 mL.
Exemplary formulation 4-tablet: the compounds described herein, or pharmaceutically acceptable salts thereof, can be mixed as a dry powder with a dry gelatin binder in a weight ratio of about 1. A small amount of magnesium stearate may be added as a lubricant. The mixture is formed into tablets of 450 to 900mg (150 to 300mg of active compound) in a tablet press.
Exemplary formulation 5-injection: the compound described herein, or a pharmaceutically acceptable salt thereof, may be dissolved or suspended in a buffered sterile saline injectable aqueous medium at a concentration of about 5mg/mL.
Exemplary formulation 6-tablet: the compounds described herein, or pharmaceutically acceptable salts thereof, can be mixed as a dry powder with a dry gelatin binder in a weight ratio of about 1. A small amount of magnesium stearate may be added as a lubricant. The mixture is formed into tablets of 90 to 150mg (30 to 50mg of active compound per tablet) in a tablet press.
Exemplary formulation 7-tablet: the compounds described herein, or pharmaceutically acceptable salts thereof, can be mixed as a dry powder with a dry gelatin binder in a weight ratio of about 1. A small amount of magnesium stearate may be added as a lubricant. The mixture is formed into tablets of 30 to 90mg (10 to 30mg of active compound per tablet) in a tablet press.
Exemplary formulation 8-tablet: the compounds described herein, or pharmaceutically acceptable salts thereof, can be mixed as a dry powder with a dry gelatin binder in a weight ratio of about 1. A small amount of magnesium stearate may be added as a lubricant. The mixture is formed into tablets of 0.3 to 30mg (0.1 to 10mg of active compound per tablet) in a tablet press.
Exemplary formulation 9-tablet: the compounds described herein, or pharmaceutically acceptable salts thereof, can be mixed as a dry powder with a dry gelatin binder in a weight ratio of about 1. A small amount of magnesium stearate may be added as a lubricant. The mixture is formed into tablets of 150 to 240mg (50 to 80mg of active compound per tablet) in a tablet press.
Exemplary formulation 10-tablet: the compounds described herein, or pharmaceutically acceptable salts thereof, can be mixed as a dry powder with a dry gelatin binder in a weight ratio of about 1. A small amount of magnesium stearate may be added as a lubricant. The mixture is formed into tablets of 270 to 450mg (90 to 150mg of active compound per tablet) in a tablet press.
The injected dose levels range from about 0.1 mg/kg/hr to at least 10 mg/kg/hr over a period of about 1 hour to 120 hours, especially over 24 to 96 hours. A pre-load bolus of about 0.1mg/kg to about 10mg/kg or greater may also be administered to achieve a sufficient steady-state level. For human patients of 40 to 80kg, the total maximum dose is desirably not more than about 2 g/day.
For the prevention and/or treatment of long-term disorders, the treatment regimen is usually extended to many months or years, and therefore oral administration is preferred in view of patient convenience and tolerability. For oral administration, oral doses from 1 to 5 times per day, especially from 2 to 4 times per day and typically 3 times per day are representative regimens. Using these dosing regimens, about 0.01 to about 20mg/kg of a compound of the invention is provided per dose, with preferred doses each providing about 0.1 to about 10mg/kg, and especially about 1 to about 5mg/kg.
The transdermal dose is typically selected to provide similar or lower blood levels than the injected dose.
When used to avoid development of CNS disorders, administration of the above dosage levels of the compounds provided herein to a subject at risk of developing the condition is typically under the recommendation and supervision of a physician. Subjects at risk of developing a particular disorder generally include those with a family history of the disorder or those determined to be particularly susceptible to developing the disorder by genetic testing or screening.
Methods of treatment and use
The compounds of the present invention, e.g., compounds of formula (A), (I-63) or (I-67), and pharmaceutically acceptable salts thereof, as described herein, are generally designed to modulate NMDA function and thus act as oxysterols for the treatment and prevention of, e.g., CNS-related disorders in a subject. In some embodiments, the compounds described herein, e.g., compounds of formula (A), (I-63), or (I-67), and pharmaceutically acceptable salts thereof, as described herein, are generally designed to penetrate the blood-brain barrier (e.g., are designed to be transported across the blood-brain barrier). Modulation, as described herein, refers to, for example, inhibition or enhancement of NMDA receptor function. In certain embodiments, a compound described herein, e.g., a compound of formula (A), (I-63), or (I-67), or a pharmaceutically acceptable salt thereof, acts as a Negative Allosteric Modulator (NAM) of NMDA receptor function and inhibits NMDA receptor function. In certain embodiments, a compound described herein, e.g., a compound of formula (A), (I-63), or (I-67), or a pharmaceutically acceptable salt thereof, acts as a Positive Allosteric Modulator (PAM) of NMDA receptor function and enhances NMDA receptor function. In certain embodiments, a compound described herein, e.g., a compound of formula (A), (I-63), or (I-67), or a pharmaceutically acceptable salt thereof, blocks or reduces the enhancement or inhibition of NMDA receptor function by a naturally occurring substrate. Such compounds do not act as Negative Allosteric Modulators (NAMs) or Positive Allosteric Modulators (PAMs) of NMDA receptor function-these compounds may be referred to as Neutral Allosteric Ligands (NALs). In some embodiments, the disease is cancer. In some embodiments, the disease is diabetes. In some embodiments, the disease is a disorder of sterol synthesis. In some embodiments, the disease is a Gastrointestinal (GI) disorder, e.g., constipation, irritable Bowel Syndrome (IBS), inflammatory Bowel Disease (IBD) (e.g., ulcerative colitis, crohn's disease), a structural disorder affecting the gastrointestinal tract, an anal disorder (e.g., hemorrhoids, internal hemorrhoids, external hemorrhoids, anal fissure, perianal abscess, anal fistula), colonic polyps, cancer, or colitis. In some embodiments, the disease is inflammatory bowel disease.
Exemplary conditions associated with NMDA-regulation include, but are not limited to, gastrointestinal (GI) disorders, e.g., constipation, irritable Bowel Syndrome (IBS), inflammatory Bowel Disease (IBD) (e.g., ulcerative colitis, crohn's disease), structural disorders affecting the gastrointestinal tract, anal disorders (e.g., hemorrhoids, internal hemorrhoids, external hemorrhoids, anal fissures, perianal abscesses, anal fistulas), colonic polyps, cancer, colitis, and CNS disorders, e.g., disorders described herein.
Exemplary conditions associated with NMDA-modulation (e.g., CNS disorders) include, but are not limited to, adjustment disorders, anxiety disorders (including obsessive-compulsive disorders, post-traumatic stress disorders, social phobias, generalized anxiety disorder), cognitive disorders (including alzheimer's disease and other forms of dementia including cortical basal dementia-progressive supranuclear palsy, frontotemporal dementia, primary progressive aphasia, parkinson's disease dementia, and lewy body dementia), separation disorders, eating disorders, mood disorders (including depression (e.g., post-partum), bipolar disorders, dysthymic disorders, suicidal tendencies), schizophrenia or other psychotic disorders (including schizoaffective disorders), sleep disorders (including insomnia), substance abuse-related disorders, personality disorders (including obsessive-compulsive personality disorders), autism spectrum disorders (including proteins associated with Shank family (e.g., those associated with mutations in Shank 3), neurodevelopmental disorders (including Rett syndrome), multiple sclerosis, sterol synthesis disorders, smith-Lemli-optitz syndrome, pain (including acute pain, chronic pain, and neuropathic pain), epilepsy (including status epilepticus and monogenic forms of epilepsy, such as Dravet disease, multiple sclerosis nodular (TSC), and infantile spasm, stroke, subarachnoid hemorrhage, intracerebral hemorrhage, cerebral ischemia, traumatic brain injury, attention deficit disorder (including Huntington's disease and Parkinson's disease), attention deficit disorder (ADHD), and cerebral ischemia, attention deficit hyperactivity disorder, metabolic encephalopathy (including phenylketonuria), postpartum psychosis, syndromes associated with high titers of anti-NMDA receptor antibodies (including anti-NMDA receptor encephalitis), neurodegenerative disorders, neuroinflammation, neuropsychiatric lupus, niemann-Pick C disorder, and tinnitus.
In certain embodiments, a compound of the invention, e.g., a compound described herein, e.g., a compound of formula (A), (I-63), or (I-67), or a pharmaceutically acceptable salt thereof, is useful for inducing sedation or anesthesia.
In certain embodiments, the compounds described herein, or pharmaceutically acceptable salts thereof, are useful for treating or preventing adjustment disorders, anxiety disorders (including obsessive-compulsive disorders, post-traumatic stress disorder, social phobia, generalized anxiety disorder), cognitive disorders (including alzheimer's disease and other forms of dementia including corticobasal dementia-progressive supranuclear palsy, frontotemporal dementia, primary progressive aphasia, parkinson's disease dementia, and lewy body dementia), separation disorders, eating disorders, mood disorders (including depression (e.g., postpartum depression), bipolar disorders, dysthymic disorders, suicidal tendencies), schizophrenia or other psychotic disorders (including schizoaffective disorders), sleep disorders (including insomnia), substance abuse-related disorders, personality disorders (including obsessive-compulsive personality disorders), autism spectrum disorders (including disorders associated with the Shank family of proteins (e.g., those associated with mutations in Shank 3), neurodevelopmental disorders (including Rett syndrome), multiple sclerosis, sterol synthesis disorders, smith-Lemli-optitz syndrome, pain (including acute pain, chronic pain, and neuropathic pain), epilepsy (including status epilepticus and monogenic forms of epilepsy, such as Dravet's disease, multiple sclerosis (TSC), and infantile spasms), stroke, subarachnoid hemorrhage, intracerebral hemorrhage, cerebral ischemia, traumatic brain injury, movement disorders (including huntington's disease and parkinson's disease) attention deficit disorder, epilepsy, central nervous system disorders (including central nervous system disorder, and epilepsy, attention deficit hyperactivity disorder, metabolic encephalopathy (including phenylketonuria), postpartum psychosis, syndromes associated with high titers of anti-NMDA receptor antibodies (including anti-NMDA receptor encephalitis), neurodegenerative diseases, neuroinflammation, neuropsychiatric lupus, niemann-Pick C disorder, and tinnitus.
In certain embodiments, the compounds described herein, or pharmaceutically acceptable salts thereof, are useful for treating or preventing an adjustment disorder, an anxiety disorder (including obsessive-compulsive disorder, post-traumatic stress disorder, social phobia, generalized anxiety disorder), a cognitive disorder (including alzheimer's disease and other forms of dementia including corticobasal dementia-progressive supranuclear palsy, frontotemporal dementia, primary progressive aphasia, parkinson's disease dementia, and lewy body dementia), a substance abuse-related disorder, a separation disorder, an eating disorder, a mood disorder (including depression (e.g., postpartum depression), bipolar disorder, dysthymic disorder, suicidal tendency), schizophrenia or other psychotic disorder (including schizoaffective disorder), a personality disorder (including obsessive-compulsive personality disorder), an autism spectrum disorder (including those associated with mutations in Shank family proteins (e.g., shank 3)), or a postpartum psychosis.
In certain embodiments, the compounds described herein, or pharmaceutically acceptable salts thereof, are useful for treating or preventing neurodevelopmental disorders (including Rett syndrome), multiple sclerosis, sterol synthesis disorders, smith-Lemli-optiz syndrome, pain (including acute pain, chronic pain, and neuropathic pain), epilepsy (including status epilepticus and monogenic forms of epilepsy, such as Dravet disease, multiple sclerosis (TSC), and infantile spasms), stroke, subarachnoid hemorrhage, intracerebral hemorrhage, cerebral ischemia, traumatic brain injury, movement disorders (including huntington's disease and parkinson's disease) attention deficit disorder, attention deficit hyperactivity disorder, metabolic encephalopathy (including phenylketonuria), syndromes associated with high titers of anti-NMDA receptor antibodies (including anti-NMDA receptor encephalitis), neurodegenerative diseases, neuroinflammation, neuropsychiatric lupus, niemann-Pick C disorder, or tinnitus.
In certain embodiments, a compound described herein, or a pharmaceutically acceptable salt thereof, is useful for treating or preventing obsessive-compulsive disorder, depression, neuropsychiatric lupus, or schizophrenia.
In certain embodiments, a compound described herein, or a pharmaceutically acceptable salt thereof, is useful for treating or preventing huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, alzheimer's disease, dementia, parkinson's disease, ataxia, fragile-X syndrome, tourette's syndrome, levodopa-induced dyskinesia, rett syndrome, autism spectrum disorders, or traumatic brain injury.
In certain embodiments, a compound described herein, or a pharmaceutically acceptable salt thereof, is useful for treating or preventing tinnitus, neuropathic pain, or migraine.
In certain embodiments, the compounds described herein, or a pharmaceutically acceptable salt thereof, are useful for treating or preventing acute liver failure or glycine encephalopathy,
in certain embodiments, a compound described herein, or a pharmaceutically acceptable salt thereof, is useful for treating or preventing seizure or hereditary epilepsy.
In some embodiments, the compounds of the present invention, e.g., a compound of formula (a), a compound of formula (I-63), or a compound of formula (I-67) as PAM for NMDA receptor function, are useful for treating or preventing disorders (e.g., CNS-related disorders) including schizophrenia or other psychotic disorders including schizoaffective disorders, sleep disorders including insomnia, autism spectrum disorders including those associated with mutations in Shank family proteins (e.g., shank 3), multiple sclerosis, movement disorders including huntington's disease and parkinson's disease, attention deficit disorder, attention deficit hyperactivity disorder, metabolic encephalopathy including phenylketonuria, post-partum psychosis, and syndromes associated with high titers of anti-NMDA receptor antibodies including anti-NMDA receptor encephalitis.
In some embodiments, a compound of the invention, e.g., NAM compound of formula (a), a compound of formula (I-63), or a compound of formula (I-67) that is an NMDA receptor function, is useful for treating or preventing disorders (e.g., CNS-related disorders) including anxiety disorders (including obsessive-compulsive disorders, post-traumatic stress disorder, social phobia, generalized anxiety disorder), mood disorders (including depression (e.g., postpartum depression), bipolar disorder, dysthymic disorder, suicidal ideation), personality disorders (including obsessive-compulsive personality disorder), neurodevelopmental disorders (including Rett syndrome), pain (including acute and chronic pain), epilepsy (including status epilepticus and monogenic forms of epilepsy, such as Dravet disease, and composite Tuberous Sclerosis (TSC)), stroke, traumatic brain injury, accommodation disorders, neuropsychiatric lupus and tinnitus.
In some embodiments, compounds of the invention, e.g., a compound of formula (a), a compound of formula (I-63), or a compound of formula (I-67) that is a PAM or NAM that functions as an NMDA receptor, are useful for treating or preventing disorders (e.g., CNS-related disorders), including cognitive disorders (including alzheimer's disease and other forms of dementia including corticobasal dementia-progressive supranuclear palsy, frontotemporal dementia, primary progressive aphasia, parkinson's disease dementia, and lewy body dementia), sterol synthesis disorders, and eating disorders.
In another aspect, there is provided a method of treating or preventing brain excitability in a subject susceptible to or suffering from a disorder associated with brain excitability, comprising administering to the subject an effective amount of a compound of the invention, e.g., a compound of formula (A), a compound of formula (I-63), or a compound of formula (I-67), or a pharmaceutically acceptable salt thereof.
In another aspect, the invention provides a combination of a compound of the invention, e.g., a compound of formula (A), a compound of formula (I-63), or a compound of formula (I-67), or a pharmaceutically acceptable salt thereof, with another pharmacologically active agent. The compounds provided herein may be administered as the sole active agent, or they may be administered in combination with other agents. Combination administration can be carried out by any technique apparent to those skilled in the art, including, for example, separate, sequential, simultaneous and alternating administration.
In another aspect, the invention provides a method of effecting negative allosteric modulation of an NMDA receptor in a subject comprising administering to the subject a compound described herein, e.g., a compound of formula (a), a compound of formula (I-63), or a compound of formula (I-67).
Movement disorder
Also described herein are methods for treating movement disorders. As used herein, "dyskinesia" refers to a variety of diseases and disorders associated with hyperkinetic movement disorders and associated abnormal muscle control. Exemplary movement disorders include, but are not limited to, parkinson's disease and parkinson's disease (specifically defined as bradykinesia), dystonia, chorea and huntington's disease, ataxia, levodopa-induced dyskinesia, tremor (e.g., essential tremor), myoclonus and startle, tics and Tourette's syndrome, restless legs syndrome, stiff person syndrome and gait disorder.
Tremor is an involuntary, sometimes rhythmic, contraction and relaxation of muscles that may involve oscillations or twitching of one or more body parts (e.g., hands, arms, eyes, face, head, vocal cords, torso, legs). Tremor includes hereditary, degenerative and idiopathic disorders such as hepatolenticular degeneration (Wilson's disease), parkinson's disease and essential tremor, respectively; metabolic diseases (e.g., thyroid-parathyroid disease, liver disease, and hypoglycemia); peripheral neuropathy (associated with Charcot-Marie-Tooth disease, rayderma-Levy disease, diabetes, complex regional pain syndrome); toxins (nicotine, mercury, lead, CO, manganese, arsenic, toluene); drug induced disorders (narcolepsy, tricyclic antidepressants, lithium, cocaine, alcohol, epinephrine, bronchodilators, theophylline, caffeine, steroids, valproate, amiodarone, thyroid hormone, vincristine); and psychological disorders. Clinical tremor can be classified into physiological tremor, enhanced physiological tremor, essential tremor syndrome (including classical essential tremor, essential orthostatic tremor and task and site specific tremor), dystonic tremor, parkinson's disease tremor, cerebellar tremor, holmes' tremor (i.e. erythronuclear tremor), palatal tremor, neuropathic tremor, toxic or drug-induced tremor and psychogenic tremor. Other forms of tremor include cerebellar or intention tremor, dystonic tremor, essential tremor, orthostatic tremor, parkinsonian tremor, physiological tremor, psychogenic tremor, or erythronuclear tremor.
Cerebellar tremor or intention tremor is a slow, broad tremor of the extremities that occurs after a targeted movement. Cerebellar tremor is caused by a lesion or lesion in the cerebellum resulting from, for example, a tumor, stroke, disease (e.g., multiple sclerosis, genetic degenerative disorder).
Dystonic tremor occurs in subjects affected by dystonia, a movement disorder in which sustained involuntary muscle contraction causes torsion and repetitive motion and/or pain and abnormal posture or position. Dystonic tremor can affect any muscle of the body. Dystonic tremor occurs irregularly and can be usually alleviated through total rest.
Essential tremor or benign essential tremor is the most common type of tremor. Some of essential tremor can be mild and non-progressive, and can be slowly progressive, starting on one side of the body but attacking both sides within 3 years. The hands are most commonly affected, but may also involve the head, voice, tongue, legs, and torso. Tremor frequency may decrease with age, but severity may increase. Highly excited mood, stress, fever, body exhaustion or hypoglycemia can trigger tremor and/or increase its severity. Symptoms typically evolve over time and may be visible and persistent after an attack.
Orthostatic tremor is characterized by rapid (e.g., greater than 12 Hz) rhythmic muscle contractions of the legs and trunk that occur immediately after standing. Cramps are felt in the thighs and legs and the patient can shake uncontrollably when asked to stand in one place. Orthostatic tremor may occur in patients with essential tremor.
Parkinsonian tremor is caused by damage to structures controlling movement within the brain. Parkinsonian tremor is usually a precursor to parkinson's disease and is often viewed as a "rolling pill" action of the hands, which can also affect the chin, lips, legs, and torso. Onset of parkinsonian tremor usually begins after the age of 60. Motion begins on one limb or body side and may progress to include the other side.
Physiological tremor can occur in normal subjects and is not clinically significant. It is found in all voluntary muscle groups. Physiological tremor may be caused by some drugs, alcohol withdrawal, or medical conditions including overactive thyroid and hypoglycemia. Tremor typically has a frequency of about 10 Hz.
Psychological or hysterical tremor may occur at rest or during postural or kinetic movement. Patients with psychogenic tremor may suffer from a transformation disorder or another psychiatric disorder.
Erythrocytic tremor is characterized by gross, slow tremor that can occur at rest, in certain postures, and deliberately. Tremor is associated with a disorder affecting the red nucleus of the classical unusual stroke of the midbrain.
Parkinson's disease affects the dopamine-producing nerve cells in the brain. Symptoms include muscle stiffness, tremors and changes in speech and gait. Parkinson's disease is characterized by tremor, bradykinesia, rigidity, and postural instability. Parkinson's disease shares the symptoms found in parkinson's disease, but is a complex of symptoms rather than a progressive neurodegenerative disease.
Dystonia is a movement disorder characterized by persistent or intermittent muscle contractions that cause abnormal, usually repetitive movements or postures. Dystonic motion can be patterned, torsional, and can be tremor. Dystonia is usually initiated or exacerbated by voluntary action and is associated with overflow-type muscle activation.
Chorea is a neurological disorder that usually affects the shoulders, hips, and face and is characterized by abrupt involuntary movements.
Huntington's disease is a hereditary disease that causes progressive debilitating nerve cells in the brain. Symptoms include uncontrolled motion, clumsiness, and balance problems. Huntington's disease can interfere with walking, speaking, and swallowing.
Ataxia refers to the loss of complete control of body movement and can affect finger, hand, arm, leg, flesh, speech, and eye movements.
Myoclonus and startle are responses to sudden and accidental stimuli, which may be auditory, tactile, visual or vestibular.
Tics are involuntary movements, usually sudden, transient, repetitive, but not rhythmic in onset, that usually mimic normal behavior and usually occur outside the background of normal activity. Tics can be classified as motor tics or vocal tics, which are associated with motor movements and vocal tics which are associated with voice. Tics may be characterized as simple or complex. For example, simple motor tics involve only a few muscles that are limited to a particular body part.
Tourette's syndrome is an inherited neuropsychiatric disorder that occurs in childhood and is characterized by multiple types of motor tics and at least one vocal tic.
Restless leg syndrome is a neurosensory motor disorder characterized by the irreversible movement of the leg at rest.
Stiff body syndrome is a progressive movement disorder characterized by involuntary cramps and muscle stiffness, usually involving the waist and legs. A straight leg gait with lumbar hyperconvexity is usually induced. Characteristic abnormalities in EMG recordings and continuous action unit activity of paraspinal muscles are often observed. Variants include "stiff limb syndrome" which produces focal stiffness, often affecting remote legs and feet.
Gait disorders refer to abnormalities in the way or style of walking that arise from neuromuscular, arthritis, or other physical changes. Gait is classified according to the system responsible for abnormal locomotion and includes hemiplegic, biplegic, neuropathic, myopathic, parkinson, choreopathic, ataxia and sensory gait.
Mood disorders
The invention also provides methods for treating a mood disorder such as clinical depression, postpartum depression or postpartum depression, perinatal depression, atypical depression, depressive depression, psychotic major depression, dysthymia, seasonal affective disorder, dysthymia, major depressive disorder, depressive personality disorder, recurrent brief depressive disorder, minor depressive disorder, bipolar disorder or manic depression, depression resulting from a chronic medical condition, treatment resistant depression, refractory depression, suicidal ideation, or suicidal behavior.
Clinical depression is also known as major depressive disorder, major Depressive Disorder (MDD), major depressive disorder, unipolar depression, unipolar disorder, and recurrent depression, and refers to a mental disorder characterized by severe and persistent depressed mood with low self-esteem and loss of interest in or inability to appreciate pleasure in activities that are usually liked. Some individuals with clinical depression have difficulty sleeping, losing weight, and often feel agitated and irritable. Clinical depression affects the perception, thought, and behavior of a subject and can lead to a variety of emotional and physical problems. A subject with clinical depression may have the distress of performing daily activities and feel like a living being without loveliness.
Postpartum depression (PND) is also known as postpartum depression (PPD) and refers to the type of clinical depression that affects women after childbirth. Symptoms may include sadness, fatigue, changes in sleep and eating habits, decreased libido, crying episodes, anxiety, and irritability. In some embodiments, the PND is a treatment resistant depression (e.g., a treatment resistant depression as described herein). In some embodiments, the PND is a refractory depression (e.g., a refractory depression described herein).
In some embodiments, the subject with PND also experiences depression or symptoms of depression during pregnancy. This depression is referred to herein as perinatal depression. In embodiments, a subject experiencing perinatal depression has an increased risk of experiencing PND.
Atypical Depression (AD) is characterized by emotional responsiveness (e.g., abnormal loss of interest) and aggressiveness, significant weight gain, or increased appetite. Patients with AD may also have excessive sleep or lethargy (hypersomnia), heavy limb sensations and significant social impairment due to hypersensitivity to perceived interpersonal rejection.
Depressive-type depression is characterized by loss of pleasure (loss of interest) in most or all activities, unresponsiveness to pleasurable stimuli, depressed mood more pronounced than sadness or loss, excessive weight loss, or excessive guilt.
Psychotic Major Depression (PMD) or psychotic depression refers to a major depressive episode, in particular of depressive nature, in which a subject experiences psychotic symptoms such as hallucinations and hallucinations.
Dysthymia refers to major depressive disorder involving motor behavior disorders and other symptoms. The subject may become mutilated and stiff and immobile or exhibit purposeless or peculiar movements.
Seasonal Affective Disorder (SAD) refers to a type of seasonal depression in which a subject has a seasonal depressive episode pattern that comes in the fall or winter.
Dysthymia refers to a condition associated with unipolar depression in which the same physical and cognitive problems are evident. It is different from major depressive disorder and tends to last longer (e.g. at least 2 years).
Dual depression refers to a rather depressed mood (dysthymia) lasting at least 2 years and separated by periods of major depression.
Depressive Personality Disorder (DPD) refers to a personality disorder characterized by depression.
Recurrent transient depression (RBD) refers to a condition in which a subject has depressive episodes about once a month, with each episode lasting 2 weeks or less and usually less than 2-3 days.
Minor depressive disorder or mild depression refers to depression with at least 2 symptoms present for up to 2 weeks.
Bipolar disorder or manic depression causes extreme mood swings including mood high points (mania or hypomania) and low points (depression). At various times of mania, the subject may feel or exhibit abnormal distraction, vigor, or irritability. It will usually make a decision with negligible consequences. The need for sleep is typically reduced. During various periods of depression, there may be crying, poor vision and negative human vision with humans. The risk of suicidal to those with the disorder was up to 6% or more over 20 years, while self-disability occurred in 30-40%. Other mental health problems such as anxiety and substance use disorders are often associated with bipolar disorder.
Depression caused by chronic medical conditions refers to depression caused by chronic medical conditions such as cancer or chronic pain, chemotherapy, chronic stress, and the like.
Treatment-resistant depression refers to a condition in which a subject has been treated for depression but the symptoms have not improved. For example, antidepressants or psychological counseling (psychotherapy) do not alleviate the symptoms of depression in subjects with treatment-resistant depression. In some cases, the symptoms of a subject with treatment-resistant depression improve, but recur. Refractory depression occurs in patients with depression who are resistant to standard pharmacological treatments, including tricyclic antidepressants, MAOI, SSRI, and dual and triple uptake inhibitors and/or anxiolytics, as well as non-pharmacological treatments such as psychotherapy, electroconvulsive therapy, vagal stimulation, and/or transcranial magnetic stimulation.
Suicidal ideation, suicidal behavior refer to the tendency of a subject to commit suicide. Suicidal ideation is related to suicidal thoughts or unusual suicidal bias. Suicidal ideation ranges from, for example, instantaneous ideas to broad ideas, detailed plans, role-playing, incomplete attempts vary greatly. Symptoms include talking about suicide, obtaining a method of committing suicide, being isolated from the world, being pre-occupied by death, feeling stranded or desperate to a situation, increasing use of alcohol or drugs, doing risky or self-destructive things, faring away from humans as if ever.
Symptoms of depression include persistent anxiety or sadness, feelings of helplessness, despair, pessimism, worthlessness, low energy, restlessness, sleep difficulties, insomnia, irritability, fatigue, athletic challenges, loss of interest in happy activities or hobbies, lack of concentration, loss of energy, low self-esteem, lack of positive thoughts or plans, excessive sleep, overeating, loss of appetite, insomnia, self-disability, suicidal thoughts, and suicide attempts. The presence, severity, frequency, and duration of symptoms may vary on an individual basis. Symptoms of depression and its relief can be determined by a physician or psychologist (e.g., via mental state examination).
Anxiety disorder
Provided herein are methods of treating anxiety disorders. Anxiety disorders are a general term that includes several different forms of abnormal and pathological fear and anxiety. Current psychiatric diagnostic criteria are capable of distinguishing between a variety of anxiety disorders.
Generalized anxiety disorder is a common chronic condition characterized by: the anxiety state is persistent and cannot be focused on any one target or situation. Patients with generalized anxiety experience non-specific persistent fear and annoyance, and become overly concerned with everyday objects. Generalized anxiety disorder is the most common anxiety disorder affecting older adults.
In panic disorders, people suffering from temporary episodes of intense terrorism and anxiety often develop tremors, shakes, confusion, dizziness, nausea, dyspnea. APA defines these panic attacks as sudden occurrences of fear or discomfort, peaking in less than ten minutes, can last for several hours, and can be triggered by stress, fear, or even exercise; however, the specific etiology is not always evident. In addition to recurrent accidental panic attacks, the diagnosis of panic disorder requires that the attacks have long-term consequences: annoyance to the possibility of onset, persistent fear of onset in the future, or significant changes in onset-related behavior. Accordingly, a person suffering from a panic disorder experiences symptoms even beyond the scope of a particular panic attack. Often, panic patients notice normal changes in the heartbeat so that they think that their heart is ill, or they are about to develop another panic attack. In some cases, during panic attacks, awareness of the human functioning is heightened (hypervigilant), wherein any perceived physiological changes are considered a potentially life-threatening illness (i.e., extreme hypothesis).
Obsessive-compulsive disorder is a type of anxiety disorder characterized primarily by repetitive obsessive-compulsive concepts (painful, persistent, and intrusive thoughts or imaginations) and obsessive-compulsive disorders (pressing to perform specific acts or habits). The OCD thought patterns can be likened to some degree as a vague, which relates to a causal relationship that is believed to be virtually nonexistent. Usually, the process is completely illogical; for example, obsessions with specific walking patterns may be employed to mitigate the obsessions threatening injury. In many cases, obsessive-compulsive disorder is completely inexplicable, simply forcing completion of a habit triggered by a nerve. In a few cases, OCD patients may only experience obsessive-compulsive sensations, with no apparent obsessive symptoms; fewer patients experience obsessive-compulsive disorder.
The single largest type of anxiety disorder is phobia, which includes all cases of fear and anxiety induced by a particular stimulus or condition. Patients typically predict the terrorist outcome from encountering the target of their fear, which may be anything from an animal to a location to a bodily fluid.
Post-traumatic stress disorder or PTSD is an anxiety disorder resulting from a traumatic experience. Post-traumatic stress can result from extreme conditions, such as fighting, rape, seizures or even serious accidents. It may also result from prolonged exposure to severe stressors, for example soldiers able to endure a single battle but unable to cope with successive battles. Common symptoms include hallucinations, evasion behavior, and depression.
Epilepsy
Epilepsy is a brain disease characterized by repeated epileptic seizures over time. Types of epilepsy may include, but are not limited to: generalized epilepsy, e.g. childhood seizures, juvenile myoclonic epilepsy, grand mal seizures on waking, west syndrome, myoclonic arrests, partial epilepsy, e.g. psychomotor epilepsy, frontal lobe epilepsy, benign focal epilepsy in childhood.
Epileptogenesis
Epileptogenesis is a progressive process by which the normal brain develops epilepsy (a chronic condition of seizures). Epileptogenesis is caused by neuronal damage that accumulates from an initial insult (e.g., status epilepticus).
Status Epilepticus (SE)
Status Epilepticus (SE) may include, for example, convulsive status epilepticus, e.g., early status epilepticus, complete status epilepticus, refractory status epilepticus, super-refractory status epilepticus; non-convulsive status epilepticus, e.g., generalized status epilepticus, complex partial status epilepticus; generalized periodic epileptiform discharges; and periodic unilateral epileptiform discharges. Convulsive status epilepticus is characterized by: convulsive status epilepticus seizures are present and may include early status epilepticus, established status epilepticus, refractory status epilepticus, and ultra-refractory status epilepticus. Early status epilepticus can be treated with first line therapy. Complete status epilepticus is characterized by: status epilepticus persists, but seizures persist despite treatment with first-line and second-line therapies. Refractory status epilepticus is characterized by: status epilepticus seizures, although treated with first-line and second-line therapy, persist and are usually under general anesthesia. Status epilepticus, which is extremely difficult to treat, is characterized by: status epilepticus seizures, although treated with first-line therapy, second-line therapy, and general anesthesia, persist for 24 hours or more.
Non-convulsive status epilepticus may include, for example, focal non-convulsive status epilepticus, e.g., complex partially non-convulsive status epilepticus, simple partially non-convulsive status epilepticus, mild non-convulsive status epilepticus, generalized non-convulsive status epilepticus, e.g., tardive non-convulsive status epilepticus, atypical non-convulsive status epilepticus, or typical unconscious non-convulsive status epilepticus.
Epileptic seizure
Seizures are physical findings or behavioral changes that occur after the onset of abnormal electrical activity in the brain. The term "seizure" is often used interchangeably with "convulsions". When the body of a person shakes rapidly and uncontrollably, it is convulsions. During convulsions, the muscles of a human repeatedly contract and relax.
Based on the type of behavior and brain activity, seizures are divided into two broad categories: generalized and partial (also called local or focal) seizures. Classification of seizure types can help physicians diagnose whether a patient has epilepsy.
The electrical impulses are distributed throughout the brain, causing generalized seizures, while the electrical impulses, in a relatively small portion of the brain, cause partial seizures (at least initially). The portion of the brain that produces the epileptic seizure is sometimes referred to as the lesion.
There are six types of generalized seizures. The most common and prominent, and therefore the most well known, is generalized convulsions, also known as seizures. In such seizures, the patient loses consciousness and often collapses. After loss of consciousness, generalized body stiffening (called the "tonic" phase of the seizure) continues for 30 to 60 seconds, then a strong spasm (the "clonic" phase) for 30 to 60 seconds, after which the patient goes to deep sleep (the "post-seizure" or post-seizure phase). During a bout, injuries and accidents may occur, such as tongue biting and urinary incontinence.
Absence attacks result in a short loss of consciousness (only a few seconds) with few symptoms. Patients (usually mostly children) typically discontinue activity and become dull. These episodes begin and end abruptly and may occur several times a day. Patients are generally unaware that they have a seizure, except that they may be aware of "lost time".
Myoclonic episodes include occasional spasms, usually on both sides of the body. Patients sometimes describe spasms as transient shocks. When episodes are intense, the episodes may cause a fall, or an object is thrown unintentionally.
Clonic seizures are repetitive, rhythmic spasms that involve both sides of the body.
Tonic seizures are characterized by muscle stiffening.
Atonic seizures involve a sudden and generalized loss of muscle tone, particularly in the arms and legs, often resulting in a fall.
Seizures described herein may include: epileptic seizures; repeated episodes of acute nature; a cluster episode; a continuous episode; an uninterrupted episode; long-term attacks; recurrent episodes; status epilepticus, e.g., refractory convulsive status epilepticus, nonconvulsive status epilepticus; refractory episodes; onset of muscle clonus; a tonic seizure; tonic-clonic seizures; simple partial seizures; a complex partial seizure; secondary generalized seizures; atypical absence episodes; absence episodes; atonic seizures; benign polycystic episode of rowland; a febrile episode; an episode of affective disorder; focal episodes; (iii) a dementia-smiling episode; generalized seizures; infantile convulsions; focal seizures; large-scale bi-directional myoclonic seizures; multifocal episodes; seizures in neonates; seizures in the night; seizures of occipital lobe; seizures after trauma; a small attack; a Sylvan episode; an episode of visual reflexivity; or withdrawal cramps. In some embodiments, the epileptic seizure is a generalized epileptic seizure associated with Dravet syndrome, lennox-Gastaut syndrome, tuberous sclerosis complex, rett syndrome, or PCDH19 Female Pediatric epilepsy.
Examples
In order that the invention described herein may be fully understood, the following examples are set forth. The synthetic and biological examples described herein are illustrative of the compounds, pharmaceutical compositions, and methods provided herein and should not be construed as limiting their scope in any way. In the following synthetic examples, the descriptions of the experimental procedures within the reaction sequence are listed in numerical order.
In some cases, the stereochemistry specified herein (e.g., assigning "R" or "S" to the C22 position of a steroid) may be specified experimentally (e.g., randomly). For example, when the absolute configuration is "S", the C22 position can be plotted in the "R" configuration. When the absolute configuration is "R", the C22 position can also be plotted in the "S" configuration. This random assignment applies to compounds 7, 13, 14, 18, 19, 22, 25, 27, 31, 37, 41, 50, 55, 60, 63, 66, 68, 73, 79, 86, 89, 91 and 99.
Materials and methods
The compounds provided herein can be prepared from readily available starting materials using the following general methods and procedures. It is to be understood that where typical or preferred process conditions (i.e., reaction temperatures, times, molar ratios of reactants, solvents, pressures, etc.) are given, other processing conditions may also be used unless otherwise indicated. Optimum reaction conditions may vary depending on the particular reactants or solvents used, but the conditions can be determined by one skilled in the art by routine optimization.
Furthermore, it will be apparent to those skilled in the art that conventional protecting groups may be required to avoid undesirable reactions with specific functional groups. The selection of suitable protecting groups for specific functional groups and suitable conditions for protection and deprotection are known in the art. Various Protecting Groups and their introduction and removal are described, for example, in t.w.greene and p.g.m.wuts, protecting Groups in Organic Synthesis, second edition, wiley, new York,1991, which are incorporated herein by reference.
The compounds provided herein can be isolated by known standard proceduresAnd purifying. Such operations include, but are not limited to, recrystallization, column chromatography, HPLC, or Supercritical Fluid Chromatography (SFC). The following schemes are provided which detail the preparation of representative pyrazoles listed herein. The compounds provided herein can be prepared by one skilled in the art of organic synthesis from starting materials and reagents that are known or commercially available. Exemplary chiral columns that can be used to separate/purify the enantiomers/diastereomers provided herein include but are not limited to,
Figure BDA0003762336660000721
AD-10、
Figure BDA0003762336660000722
OB、
Figure BDA0003762336660000723
OB-H、
Figure BDA0003762336660000724
OD、
Figure BDA0003762336660000725
OD-H、
Figure BDA0003762336660000726
OF、
Figure BDA0003762336660000727
OG、
Figure BDA0003762336660000728
OJ and
Figure BDA0003762336660000729
OK。
exemplary general procedure for preparative HPLC: column: waters Rbridge prep 10 μm C18,19 × 250mm. Mobile phase: acetonitrile, water (NH) 4 HCO 3 ) (30L of water, 24g of NH) 4 HCO 3 ,30mL NH 3 .H 2 O). Flow rate: 25mL/min
Exemplary general methods for analytical HPLC: mobile phase: a: water (10 mM) NH 4 HCO 3 ) B, the following steps: acetonitrile, gradient: 5% -95% by weight B for 1.6 or 2min; flow rate: 1.8 or 2mL/min; column: XBridge C18,4.6 × 50mm,3.5 μm @45c.
NMDA modulation
Evaluation of NMDA enhancement in mammalian cells expressing the NMDA receptor using an automated patch clamp system can be used to determine NAM activity of compounds as described below. The PAM activity of the compounds described below can be determined using a whole cell patch clamp system.
Automated patch clamp system (QPatch HTX):
in this study, HEK 293 cells stably transfected with glutamate-activated GRIN1/2A subtype channels will be used with sub-maximal NMDA concentrations (300 μ M NMDA, applied with 8 μ M glycine) to study negative allosteric modulation of test compounds. The percent enhancement results obtained with this method are shown in table 1.
Cell culture
Typically, cells will be passaged at about 80% to about 90% confluence. For electrophysiological measurements, cells will be harvested from sterile culture flasks containing culture complete medium at about 80% to 90% confluence. Cells were transferred as a suspension in PBS to QPatch 16X or QPatch HTX systems for direct transfer to centrifuge/washer.
Standard laboratory conditions: the cells were assayed at 37 ℃ for 5% CO 2 In a humid atmosphere (relative humidity about 95%).
Culture medium: cells will be maintained and passaged continuously in sterile culture flasks containing Dulbecco's modified eagle's medium and a 1.
Antibiotics: complete media as described above were supplemented with 100. Mu.g/mL hygromycin, 15. Mu.g/mL blasticidin, and 1. Mu.g/mL puromycin.
Induction of expression: 2.5. Mu.g/mL tetracycline was added 24 hours before the start of the experiment.
Dosage formulations
Dosage levels are based on the test compound provided. The vehicle was added to achieve a stock concentration of 10mM (stored at-10 ℃ to 30 ℃). Another stock solution was prepared at 1.0mM in DMSO. Details of stock use (thawing, dosage formulation) will be recorded in the raw data. The time period of stock solution use will be detailed in the report.
Test Compound concentration
Dosage levels are in accordance with the test compound provided. The vehicle was added to achieve a stock concentration of 10mM (stored at-10 ℃ to 30 ℃). Another stock solution was prepared at 1.0mM in DMSO. Details of stock use (thawing, dosage formulation) will be recorded in the raw data. The time period of stock solution use will be detailed in the report.
One test concentration of 1.0 μ M will be tested.
All test solutions will be prepared by diluting stock solutions with either Mg-free baths alone or magnesium-free baths containing NMDA (300 μ M) and glycine (8.0 μ M) shortly before electrophysiological experiments and kept at room temperature (19 ℃ to 30 ℃) at the time of use. DMSO 0.1% was used as a carrier.
Preparation frequency: for each concentration tested, a new test compound solution will be prepared daily.
Stability of the dosage formulation: all preparation times will be recorded in the raw data. Any observations regarding the instability of the test compounds will be mentioned in the raw data.
Storage of the dosage formulation: on the day of the experiment, the dosage formulations will remain at room temperature (19 ℃ to 30 ℃) when used.
Bath lotion
To prepare the experiments and form the gigaseal, the following standard baths will be used:
sodium chloride: 137mM; potassium chloride: 4mM; calcium chloride: 1.8mM; magnesium chloride: 1mM; HEPES (high efficiency particulate air): 10mM; d-glucose: 10mM; cremophor: 0.02 percent; pH (NaOH): 7.4
The 1x bath was prepared by diluting the 10x bath and the 100x glucose solution without glucose with water at least every 7 days. Both stock solutions were prepared prior to the start of the experiment in this study and stored at 1 ℃ to 9 ℃ (10 x bath solution) or-10 ℃ to-30 ° (100 x glucose solution). The batch number of the bath used in the experiment will be recorded in the raw data. When used, the 1 × bath will be maintained at room temperature (19 deg.C to 30 deg.C). When not in use, the 1 × bath will be stored at 1 deg.C to 9 deg.C.
After forming the gigaseal, the following magnesium-free baths will be used:
sodium chloride: 137mM; potassium chloride: 4mM; calcium chloride; 2.8mM; HEPES (high efficiency particulate air): 10mM; d-glucose: 10mM; cremophor: 0.02 percent; pH (NaOH): 7.4
The Mg-free bath was prepared as a 1x solution and stored at 1 ℃ to 9 ℃. It will be freshly prepared at least once every 10 days.
Intracellular solutions
The 1x intracellular solution, which had been prepared prior to the start of the experiment in this study, was thawed daily from a frozen 1x intracellular solution, aliquoted and stored at-10 ℃ to-30 ℃. In use, the 1x intracellular solution will be maintained at room temperature (19 ℃ to 30 ℃). The remaining 1x intracellular solution will be stored in a refrigerator (1 ℃ to 9 ℃). The 1x intracellular solution will include the components listed below:
potassium chloride: 130mM; magnesium chloride: 1mM; mg-ATP:5mM; HEPES (high efficiency particulate air): 10mM; EGTA:5mM; pH (KOH): 7.2
Cell processing
For this study, cells will be perfused continuously with NMDA/glycine, test compound or test compound/NMDA/glycine.
In each case, a pre-wash step with test compound was performed for at least 30 seconds between applications. For detailed information, see table a below.
Each experimental type will be analyzed in at least n =3 isolated cells. NMDA and glycine stock solutions were prepared prior to the start of the experiment in this study and stored frozen (-10 ℃ to-30 ℃) until the day of the experiment. Shortly before the electrophysiological experiments, frozen stock solutions were thawed and diluted.
Comparison: the effects of vehicle (0.1% DMSO) and D- (-) -2-amino-5-phosphonopentanoic acid (AP-5) (100. Mu.M) were measured in three cells every other week to ensure successful expression of NMDA receptor.
A50 mM AP-5 stock solution was prepared prior to the start of the experiment in this study, aliquoted and stored frozen (-10 ℃ to-30 ℃) until the day of the experiment. Shortly before electrophysiological experiments frozen stock solutions were thawed and then diluted in Mg-free baths containing NMDA (300 μ M) and glycine (8.0 μ M) to give a final perfusion concentration of 100 μ M.
Experimental procedure
Cells were transferred as a suspension in serum-free medium to the QPatch HTX system and maintained in a cell storage tank/blender during the experiment. All solutions applied to the cells (including intracellular solutions) will be maintained at room temperature (19 ℃ to 30 ℃).
During the sealing process, the standard baths described above will be used. All solutions applied to the cells (including pipette solutions) will be maintained at room temperature (19 ℃ to 30 ℃). After a Gigaohm seal is formed between the patch electrode and transfected HEK293 cells alone, only the Mg-free bath will be perfused and the cell membrane will rupture to ensure that electricity enters the cell interior (whole cell patch configuration). The inward current was measured 5 seconds after 300. Mu.M NMDA (and 8.0. Mu.M glycine) was applied to the patch-clamp cells. Throughout the experiment, cells will be held at a holding potential of-80 mV by a voltage clamp.
To analyze the test compounds, the NMDA receptor will be stimulated by 300 μ M NMDA and 8.0 μ M glycine in combination with the test compounds described below. A thirty-second pre-wash step with test compound will be performed between the two applications.
Table a: an application protocol; testing the use dependence of Compounds
Figure BDA0003762336660000751
Figure BDA0003762336660000761
Table B: an application protocol; control experiment
Figure BDA0003762336660000762
Whole-cell patch clamp for mammalian cells (Ionworks Barracuda (IWB))
The whole-cell patch clamp technique was used to study the positive allosteric regulatory activity of test compounds on the GlunN1/GluN2A and GluN2B glutamate receptors expressed in mammalian cells. EC (EC) 50 And E max The data are shown in table 1.
HEK293 cells were transformed with adenovirus 5DNA and transfected with cDNA encoding the human GRIN1/GRIN2A gene. Stable transfectants were selected using G418 and anti-Zeocin genes incorporated into the expression plasmid, and selection pressure was maintained with G418 and Zeocin in the medium. Cells were cultured in Duchen's modified Eagle Medium/nutrient mixture (D-MEM/F-12) supplemented with 10% fetal bovine serum, 100. Mu.g/ml penicillin G sodium, 100. Mu.g/ml streptomycin sulfate, 100. Mu.g/ml Zeocin, 5. Mu.g/ml blasticidin and 500. Mu.g/ml G418.
Test article effects were evaluated in an 8-point concentration-response mode (4 for duplicate wells/concentration). All test and control solutions contained 0.3% DMSO and 0.01%
Figure BDA0003762336660000763
EL (C5135, sigma). The test article formulations were loaded in 384-well compound plates using an automated liquid handling system (SciClone ALH3000, caliper lifesciences). Measurements were performed using the Ion Works Barracuda platform according to the following procedure:
electrophysiological operation:
a) Intracellular solution (mM): 50mM CsCl,90mM CsF,2mM MgCl 2 5mM EGTA,10mM HEPES. Adjusted to pH 7.2 with CsOH.
b) Extracellular solution, HB-PS (composition, in mM): naCl,137; KCl,1.0; caCl 2 5, 5; HEPES,10; glucose, 10; adjusted to pH 7.4 with NaOH (refrigerated before use).
c) Holding potential: -70mV, potential during agonist/PAM application: -40mV.
Recording operation:
a) Extracellular buffer was loaded in the wells of the PPC plate (11. Mu.L per well). The cell suspension was titrated into the wells of the PPC planar electrode (9 μ Ι per well).
b) Whole cell recordings were created via patch punch-through, where membrane currents were recorded by an on-board (on-board) patch-clamp amplifier.
c) Two recordings (scans) were performed. First, during pre-application of the individual test articles (duration of pre-application-5 min), and second, co-application of test article and agonist (EC) 20 L-glutamic acid and 30 μ M glycine), thereby testing the positive modulating effect of the test article.
Administration of the test article: the first pre-application consisted of the addition of 20. Mu.L of a 2 Xconcentration solution of the test article, and the second consisted of the addition of 20. Mu.L of a 1 Xconcentration solution of the test article and 10. Mu.L/s of agonist (total application time of 2 seconds).
Enhancement of channels by Positive Allosteric Modulators (PAM)
The enhancement of the channel by a Positive Allosteric Modulator (PAM) will be calculated as
% activation = (I) PAM /I EC10-30 )x100%-100%
Wherein I PAM Will be L-glutamic acid EC 10-30 The current induced in the presence of various concentrations of the test substance, I EC20 Will be with L-glutamic acid EC 20 The average current induced.
The PAM concentration-response data will conform to the equation of the form:
% activation =% L-glutamic acid EC 20 + { (% MAX-% L-glutamic acid EC 20 ) /[1+ ([ test ]]/EC 50 ) N ]},
Wherein [ test]Will be the concentration of PAM (test article), EC 50 Will be the concentration of PAM that produces half-maximal activation, N will be the Hill coefficient,% L-glutamic acid EC 20 Will be L-glutamic acid EC 20 Percentage of induced current,% MAX is with L-glutamic acid EC 20 Highest co-approvedPercent current for dose PAM activation,% activation would be with L-glutamic acid EC at each PAM concentration 10-30 Percentage of induced current.
The maximum amplitude of the induced current is measured and defined as the Peak Current Amplitude (PCA).
Abbreviations
PCC: pyridinium chlorochromate; t-BuOK: potassium tert-butoxide; 9-BBN: 9-borabicyclo [3.3.1]Nonane; pd (t-Bu) 3 P) 2 : bis (tri-tert-butylphosphino) palladium (0); acCl: acetyl chloride; i-PrMgCl: isopropyl magnesium chloride; TBSCl: tert-butyl (chloro) dimethylsilane; (i-PrO) 4 Ti: titanium tetraisopropoxide; BHT:2, 6-di-tert-butyl-4-methylphenoxide; me: a methyl group; i-Pr: isopropyl group; t-Bu: a tertiary butyl group; ph: a phenyl group; et: an ethyl group; bz: a benzoyl group; bzCl: benzoyl chloride; csF: cesium fluoride; DCC: dicyclohexylcarbodiimide; DCM: dichloromethane; DMAP: 4-dimethylaminopyridine; DMP: dess-Martin periodinane; etMgBr: ethyl magnesium bromide; etOAc: ethyl acetate; TEA: triethylamine; alaOH: alanine; boc: tert-butoxycarbonyl group. Py: pyridine; TBAF: tetra-n-butylammonium fluoride; THF: tetrahydrofuran; TBS: tert-butyldimethylsilyl group; TMS: a trimethylsilyl group; TMSCF 3 : (trifluoromethyl) trimethylsilane; ts: a p-toluenesulfonyl group; bu: a butyl group;
Ti(OiPr) 4 : titanium tetraisopropoxide; LAH: lithium aluminum hydride; LDA: lithium diisopropylamide; h, liOH 2 O: lithium hydroxide hydrate; and (3) MAD: methylaluminum bis (2, 6-di-tert-butyl-4-methylphenoxide); meCN: acetonitrile; NBS: n-bromosuccinimide; na (Na) 2 SO 4 : sodium sulfate; na (Na) 2 S 2 O 3 : sodium thiosulfate; PE: petroleum ether; meCN: acetonitrile; meOH: methanol; boc: a tert-butoxycarbonyl group; MTBE: methyl tert-butyl ether; the DIAD: diisopropyl azodicarboxylate; sat.: saturation; aq.: water-based; hr/hrs: hour/hour; min/mins: minutes per minute.
Example 1: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -10, 13-dimethyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybut-2-yl) -3- (trifluoromethyl) -2,3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (1)
Figure BDA0003762336660000781
Figure BDA0003762336660000791
1. To a solution of TBAF (3.04mL, 1M in THF, 3.04mmol, aldrich) in THF (100 mL) at 0 deg.C was added TMSCF 3 (25.8g, 182mmol) and then a solution of S-200-INT-2 (19g, 60.8mmol) in THF (100 mL) was added dropwise. The mixture was stirred at 0 ℃ for 30 minutes. TBAF (200mL, 1M in THF, 200mmol, home) was added to the mixture at 0 ℃. The mixture was stirred at 0 ℃ for a further 30mins. Adding NH to the mixture 4 Cl (100mL, sat., aq.). The mixture was concentrated in vacuo. To the residue was added PE/EtOAc (400mL, 1), the organic layer was separated and combined with two other batches (2x 10g S200-INT-2). The combined organic layers were washed with water (300 mL), brine (300 mL), and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give an oil. The residue was dissolved in DCM (150 mL) and diluted with PE (750 mL). The solution was poured into a silica gel column (500g, 100-200 mesh) and eluted with PE: DCM: etOAc = 5. The impurities were recrystallized from MeCN (250 mL) to give S200-CF3_1A (6.5 g) as a solid. The filtered form MeCN was purified by silica gel column (PE: DCM: etOAc =50 1 to 20.
Note: 200-CF3_1A and 200-CF3_1B are composed of 3 J H,CF3 (FDCS) (.j.org.chem.2015, 80, 1754 identification.
S-200-CF3_1A:
1 H NMR(400MHz,CDCl 3 )δ5.43-5.33(m,1H),4.85(s,1H),4.71(s,1H);2.49(s,2H);2.11-1.97(m,4H),1.95-1.32(m,14H),1.30-0.98(m,7H),0.59(s,3H)。
S-200-CF3_1B:
1 H NMR(400MHz,CDCl 3 )δ5.54-5.41(m,1H),4.86(s,1H),4.72(s,1H);2.78-2.65(m,1H);2.18-1.97(m,3H),1.95-1.35(m,16H),1.32-0.98(m,7H),0.59(s,3H)。
2. To a solution of S-200-CF 3-1A (8g, 20.9mmol) in THF (80 mL) was added 9-BBN dimer (5.85g, 24mmol). The mixture was stirred at 40 ℃ for 1h. The mixture was cooled to 0 ℃. EtOH (12 mL), naOH (41.8mL, 5M, aq.) and H were added dropwise to the mixture 2 O 2 (20.9mL, 10M, aq.). The mixture was stirred at 50 ℃ for 1h. After cooling, na was added to the mixture 2 SO 3 (100mL, 25%, aq.). The mixture was extracted with EtOAc (300 mL). The organic layer was separated and purified by silica gel column (PE: etOAc =10, 1 to 5) to give S-200-CF3_2A (7.1g, 85%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.42-5.32(m,1H),3.64(dd,J=3.2,10.4Hz,1H),3.37(dd,J=6.8,10.4Hz,1H),2.49(s,2H),2.32-1.92(m,4H),1.92-1.70(m,4H),1.70-1.29(m,8H),1.29-0.91(m,11H),0.71(s,3H)。
3. DMP (6.31g, 14.9 mmol) was added to a solution of S-200-CF 3-5A (3 g, 7.49mmol) in DCM (50 mL) at 25 deg.C, and after stirring for 30 minutes at 25 deg.C, the reaction mixture was saturated with NaHCO 3 Quench (100 mL) and add DCM (100 mL) and stir for 10 min. The DCM phase was separated and saturated Na was used 2 S 2 O 3 Aqueous (2 × 100 mL) wash. The combined organic layers were washed with saturated brine (2X 100 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (5-20% EtOAc in PE) to give N-004-027 \u1 (1.5g, 50%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.58-9.55(m,1H),5.38-5.36(m,1H),2.49(s,1H),2.40-2.25(m,1H),2.23-1.60(m,10H),1.53-1.20(m,9H),1.15-1.00(m,7H),0.78-0.64(m,3H)。
4. To N-004-027. Sup.1 (1.5g76 mmol) in anhydrous THF (40 mL) CsF (1.42g, 9.40mmol) was added. After stirring at 0 ℃ for 20 minutes, TMSCF was added at 0 DEG C 3 (1.33g, 9.40mmol) and stirred for 30 minutes. The color changed to light yellow. Addition of TBAF.3H 2 O (4.74g, 15.0 mmol) and stirred at 50 ℃ for 30 min. The reaction mixture was poured into ice-water (100 mL). The aqueous phase was extracted with EtOAc (2 × 100 mL). The combined organic phases were washed with saturated brine (2X 100 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated to give the isomer mixture (1.45 g, crude) as a yellow solid which was purified by flash column (0-15% EtOAc in PE) to give white solid 53 (340mg, 24%) and white solid 1 (200mg, 14%).
1:
1 H NMR(400MHz,CDCl 3 )δ5.38-5.36(m,1H),4.10-4.00(m,1H),2.49(s,2H),2.19-2.12(m,1H),2.06-1.61(m,10H),1.53-1.29(m,6H),1.27-0.98(m,10H),0.71(s,3H)。
LCMS Rt =1.121 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity,
MS 50-100 \u1 \u4 min. M for C 24 H 33 F 6 O[M+H-H 2 O] + 451, found value 451.
1:
1 H NMR(400MHz,CDCl 3 )δ5.38-5.36(m,1H),4.10-4.00(m,1H),2.49(s,2H),2.19-2.12(m,1H),2.06-1.61(m,10H),1.53-1.29(m,6H),1.27-0.98(m,10H),0.71(s,3H)。
LCMS Rt =1.121 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity,
MS 50-100 \u1 \u4 min. M for C 24 H 33 F 6 O[M+H-H 2 O] + 451, found value 451.
Example 2: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -3- (methoxymethyl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (2)
Figure BDA0003762336660000811
Figure BDA0003762336660000821
1. To a suspension of N-4 (50g, 157mmol) in dry methanol (500 mL) at 20 ℃ was added dry TsOH (2.84g, 15.7 mmol) in portions. The mixture was warmed to 60 ℃ and stirred for 1h. Reaction mixture with Et 3 N (1.58g, 15.7 mmol) was quenched and stirred for an additional 30 minutes. The precipitated solid was filtered off, washed with methanol (250 mL) and air dried to give N-4-1_2 (51g, 90%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.18(s,3H),3.14(s,3H);2.54-2.48(m,1H);2.10-2.00(m,4H);1.95-1.75(m,2H),1.65-1.50(m,7H),1.48-0.80(m,11H),0.78-0.75(m,4H),0.59(s,3H)。
2. At N 2 At 20 ℃ to Ph 3 To a suspension of PMeBr (75g, 210mmol) in dry THF (500 mL) was added t-BuOK (23.5g, 210mmol) in portions. The mixture turned dark clear and was stirred for 30mins at 20 ℃. N-4-1_2 (51g, 140mmol) was then added. The mixture was warmed to 40 ℃ and stirred for 1h. The reaction mixture was cooled and poured in portions of NH 4 Aqueous Cl (ice) (400 mL). Separating the resulting mixture; the aqueous layer was extracted with THF (200 mL). The combined organic layers were used directly as a solution of N-4-1 \/3 without further purification.
3. To a solution of N-4-1_3 (50.4g, 139mmol) in THF (700 mL) at 20 deg.C was added aqueous HCl (1M, 208mL, 208mmol). The mixture was stirred at 20 ℃ for 1 hour, and a solid precipitated. Water (200 mL) was added to the mixture, and the precipitated solid was filtered off, washed with water and dried to give N-4-1 \u4 (41g, 94%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.85(s,1H),4.70(s,1H);2.38-2.25(m,3H);2.10-1.98(m,3H),1.88-1.49(m,10H),1.40-1.08(m,11H),0.97-0.72(m,2H),0.58(s,3H)。
4. At 25 ℃ inN 2 Downward Me 3 To a solution of SI (101g, 496mmol) in anhydrous THF (400 mL) was added t-BuOK (58.3g, 520mmol) portionwise and stirred for 30mins. A solution of N-4-1 \/4 (39g, 124mmol) in dry THF (300 mL) was added. The reaction mixture was warmed to 50 ℃ and stirred for 2 hours. The reaction mixture was cooled to 25 ℃ and washed with aq 4 Cl (500 mL). The aqueous phase was extracted with EtOAc (2 × 500 mL). The combined organic phases were washed with brine (2X 300 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (PE/EtOAc =20/1 to 10/1) to give N-4-3_1 (35 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.70(s,1H);2.65-2.55(m,2H);2.10-1.98(m,2H),1.92-1.49(m,13H),1.40-1.13(m,8H),0.99-0.69(m,6H),0.57(s,3H)。
5. To a solution of N-4-3_1 (35g, 647mmol) in anhydrous MeOH (500 mL) at 25 deg.C was added MeONa (57.2g, 1.06mol) and the mixture was added N 2 Stirred for 30 minutes. The reaction mixture was warmed to 70 ℃ and at N 2 The mixture was stirred at reflux for 3 hours. The reaction mixture was cooled to 25 ℃ and treated with water (500 mL). The aqueous phase was extracted with DCM (2 × 300 mL). The combined organic phases were washed with saturated brine (2X 300 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solid. The residue was purified by silica gel chromatography (PE/EtOAc =10/1 to 6/1) to give N-4-3 \u2 (25 g, impure) as a solid. The crude product was triturated at 25 ℃ in PE (250 mL) for 1h. The suspension was filtered and the filter cake was dried in vacuo to give N-4-3- \ u 2 (15g, 25%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.86(s,1H),4.72(s,1H),3.46-3.37(m,5H),2.54(s,1H),2.07-1.99(m,1H),1.89-1.52(m,15H),1.41-1.06(m,10H),0.86(s,3H);0.58(s,3H)
6. To a solution of N-4-3_2 (15g, 41.6 mmol) in anhydrous THF (200 mL) at 0 deg.C was added 9-BBN dimer (27.7g, 124mmol) and the reaction mixture was quenched with N 2 Stirring for 30mins. The reaction mixture was warmed to 50 ℃ and stirred for 1h. The reaction mixture is cooled to 0 ℃ andEtOH (50 mL) was added followed by very slow addition of NaOH (41.6 mL,5M, 208mmol) at 0 ℃. Slow addition of H 2 O 2 (23.5g, 208mmol,30% in water) while maintaining the internal temperature below 10 ℃. The mixture was warmed to 50 ℃ and stirred for an additional 1h. The reaction mixture was cooled, poured in portions into ice-water (500 mL) and filtered. The filtrate was concentrated in vacuo to afford N-4-3 \ (3) (14 g, crude) as an oil. The crude residue was used directly in the next step.
7. DMP (3.35g, 7.92mmol) was added to a mixture of N-4_3 (1g, 2.64mmol) in DCM (20 mL) at 25 ℃. The reaction mixture was warmed to 40 ℃ and stirred for 1h. The reaction mixture is treated with saturated NaHCO at pH 7-8 and below 10 deg.C 3 The aqueous solution was quenched. The suspension was filtered. The DCM phase in the filtrate was separated and saturated NaHCO was used 3 /Na 2 S 2 O 3 Aqueous solution (1, 2x50ml), brine (2 × 50 mL), washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solid. The residue was purified by flash column (0-30% EtOAc in PE) to give N-4-3_4 (0.6g, 60%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.57(s,1H),3.40-3.34(m,5H);2.38-2.28(m,1H);1.94-1.76(m,2H),1.74-1.35(m,16H),1.06-0.82(m,10H),0.73-0.64(m,5H)。
8. At 0 ℃ in N 2 Next, isopentyl magnesium bromide (4.37mL, 8.74mmol 2M in ether) was added to a solution of N-4-3-u 4 (0.6 g, 1.59mmol) in anhydrous THF (10 mL). The reaction mixture was warmed to 25 ℃ and stirred for 1 hour. Adding saturated NH to the reaction mixture 4 Aqueous Cl (50 mL). The aqueous phase was extracted with EtOAc (3 × 50 mL). The combined organic phases were washed with saturated brine (2X 50 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated to give N-4-4A (0.5 crude) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.64-3.60(m,1H),3.40-3.37(m,5H);2.02-1.79(m,3H);1.75-1.50(m,11H),1.25-1.10(m,14H),0.99-0.75(m,14H),0.70-0.64(m,4H)。
9. DMP (1.88g, 4.44mmol) was added to the flask at 25 deg.CN-4-4A (0.5 g, crude) in DCM (20 mL). The reaction mixture was warmed to 40 ℃ and stirred for 1 hour. The reaction mixture is treated with saturated NaHCO at pH 7-8 and below 10 deg.C 3 The aqueous solution was quenched. The suspension was filtered. The DCM phase was separated and saturated NaHCO was used 3 /Na 2 S 2 O 3 Aqueous solution (1, 2x50ml), brine (2 × 50 mL), washed with Na 2 SO 4 And (5) drying. Filtration and concentration in vacuo afforded N-4-4O (0.4 g, crude) as a solid, which was used directly in the next step.
NaBH is added at 25 deg.C 4 (0.340g, 8.95mmol) was slowly added to a solution of N-4-4O (0.4g, 0.895mmol) in MeOH (4 mL) and stirred for 2 hours. The aqueous phase was extracted with DCM (2 × 20 mL). The combined organic phases were washed with saturated brine (2X 20 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solid. The residue was purified by silica gel chromatography (PE/EtOAc =8/1 to 5/1) to give 35 (150 mg, impure) and 2 (130 mg, impure) as solids. 2 (130 mg, impure) was recrystallized from MeCN (3 mL) at 82 ℃ under reflux for 1 hour. The mixture was stirred and cooled to 25 ℃. The suspension was filtered and the filtrate was concentrated in vacuo to afford 2 (50mg, 12%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.63-3.61(m,1H),3.41-3.38(m,5H);2.51(s,1H);1.97-1.81(m,2H),1.71-1.31(m,15H),1.26-1.03(m,10H),0.97-0.78(m,14H),0.71-0.59(m,4H)。
LCMS Rt =1.350 min, chromatography at 2.0 min, 30-90AB, 99% purity, MS ESI C 29 H 48 O[M+H-2H 2 O] + Calculated value 413, found value 413.
Example 3: synthesis of (3S, 8R,9S,10R,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -13-methyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (3)
Figure BDA0003762336660000861
1. t-BuOH (350 mL) was added to a three-neck round bottom flask at 35 ℃ under nitrogen and stirred for 10mins under nitrogen. t-BuOK (90.5g, 807 mmol) was added to the mixture and stirred under nitrogen for 15mins. S-310-B9-1 (20g, 73.4mmol) was added to the above mixture and stirred at 35 ℃ for 1.5 hours under nitrogen. The reaction mixture was poured into 10% aqueous acetic acid (500 mL) and stirred at below 35 ℃ for 15mins. Water (500 mL) was added and the mixture was stirred for 30mins. The pH of the mixture was adjusted to 7-8 with sodium bicarbonate (500 ml) and stirred for 30 minutes. The mixture was extracted with PE (2 × 500 mL). The organic layer was separated, washed with brine (500 mL), dried over anhydrous sodium sulfate, filtered and concentrated below 35 ℃ to give S-200-N19-3 \u1 (17 g, crude) as an oil. The crude residue was used directly in the next step.
2. To a solution of 2, 6-di-tert-butyl-4-methylphenol (100g, 453mmol) in toluene (300 ml) was added AlMe dropwise at 0 DEG C 3 (113mL, 226mmol,2M in toluene). The mixture was stirred at 25 ℃ for 1hr to produce MAD. A solution of S-200-N19-3 (1: 10g,36.7 mmol) in toluene (50 mL) was added dropwise to the MAD solution at-70 ℃. After stirring at-70 ℃ for 1h, meMgBr (36.6 ml,110mmol,3M in ether) was added dropwise at-70 ℃. The resulting solution was stirred at-70 ℃ for 1 hour. The reaction mixture was quenched by saturated citric acid (400 ml) at-70 ℃. After stirring for 10min at 25 ℃, the resulting mixture was filtered and washed with EtOAc (2 × 200 ml). The combined organic layers were separated, washed with brine (2 × 200 ml), and Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (PE/EtOAc =10/1 to 5/1) to give S-200-N19-3_2 (7.6 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.45-5.40(m,1H),2.51-2.38(m,1H),2.49-2.21(m,1H),2.14-1.88(m,5H),1.86-1.77(m,2H),1.73-1.38(m,8H),1.34-1.22(m,4H),0.95-0.81(m,8H)。
3. At 40 ℃ in N 2 Downward PPh 3 To a suspension of EtBr (37.1g, 100mmol) in THF (200 mL) was added t-BuOK (11.2g, 100mmol). After stirring at 20 ℃ for 10 minutes, S-200-N19-3_2 (7.6 g, 25.1mmol) was added. The reaction mixture was stirred at 40 ℃ for 1 hour. The reaction is carried out at 0 DEG Caq.NH 4 Cl (200 mL) and extracted with EtOAc (3X 200 mL). The combined organic phases were washed with brine (200 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by Combi-flash (0% -30% etoac in PE) to give S-200-N19-3_3 (5g, 63%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.45-5.35(m,1H),5.20-5.00(m,1H),2.41-2.30(m,1H),2.29-2.12(m,3H),2.09-1.76(m,6H),1.69-1.38(m,15H),1.35-0.94(m,7H)。
4. At 0 ℃ in N 2 To a solution of S-200-N19-3 (2g, 6.35mmol) in THF (20 mL) was added 9-BNN dimer (3.09g, 12.7 mmol) next. The solution was stirred at 60 ℃ for 1 hour. After cooling to 0 ℃ a solution of EtOH (20 ml) and NaOH (12.7ml, 5M, 63.5mmol) was added very slowly. After addition, H was slowly added 2 O 2 (2.15mg, 6.35mmol,30% in water) and the internal temperature was kept below 10 ℃. The mixture was heated at 60 ℃ under N 2 Stirring was continued for 1 hour. The mixture was cooled to 30 ℃ again, water (100 mL) was added to the solution and the aqueous layer was extracted with EtOAc (100 mL). The organic layer was washed with brine (2 × 100 mL). The combined organic layers were washed with anhydrous Na 2 SO 4 Dried and purified by silica gel chromatography (PE/EtOAc = 2/1) to give S-200-N19-4 \u1 (1.6 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.45-5.35(m,1H),3.75-3.62(m,1H),2.28-2.19(m,1H),2.10-1.75(m,7H),1.71-0.97(m,19H),0.92-0.75(m,4H),0.68(s,3H)。
5. To a solution of S-200-N19-4_1 (1.6 g, 4.81mmol) in DCM (20 mL) was added silica gel (2 g) and PCC (2.07g, 9.62mmol). The mixture was stirred at 25 ℃ for 3 hours. To the mixture was added PE (50 mL). The mixture was filtered through a pad of silica gel and the solid was washed with PE/DCM (30 mL/30 mL). The mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (PE/EtOAc =10/1 to 5/1) to give S-200-N19-4_2 (1.2 g, impure) as a solid, which was recrystallized from MeCN (10 mL) at reflux to afford S-200-N19-4_2 (1.0 g, 84.0%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.40-5.35(m,1H),2.61-2.45(m,1H),2.30-2.10(m,5H),2.00-1.75(m,6H),1.70-1.10(m,14H),0.90-0.75(m,4H);0.633(s,3H)。
LCMS Rt =1.058 min, chromatography at 2.0 min, 30-90AB, purity 100% 22 H 34 [M+H-H 2 O] + Calculated value 313 of (a), measured value 313.
6. At N 2 t-BuOK (3.51g, 31.4mmol) was added to Ph at 40 ℃ below 3 PMeBr (11.1g, 31.4 mmol) in THF (50 mL). After stirring at 25 ℃ for 10 minutes, S-200-N19-4_2 (2.6g, 7.86mmol) was added. The reaction mixture was stirred at 40 ℃ for 1h. The reaction is carried out at 0 ℃ with NH 4 Aqueous Cl (100 mL) was quenched and extracted with EtOAc (2 × 100 mL). The combined organic phases were washed with brine (2X 100 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by Combi-flash (0% to 30%, etOAc in PE) to give S-200-N19-4 \u3 (2.4 g, 93%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.45-5.35(m,1H),4.86-4.83(m,1H),8.70-4.65(m,1H),2.27-2.20(m,1H),2.10-1.90(m,4H),1.89-1.50(m,11H),1.49-1.30(m,3H),1.28-1.00(m,6H),0.80-0.60(m,5H),0.59(s,3H)。
7. At 0 ℃ in N 2 Next, 9-BBN dimer (9.27g, 38.0 mmol) was added to a solution of S-200-N19-4_3 (5g, 15.2 mmol) in THF (60 mL). The solution was stirred at 60 ℃ for 1h. After cooling to 0 ℃ a solution of EtOH (60 ml) and NaOH (30.4 ml,5M, 152mmol) was added very slowly. After addition, H was slowly added 2 O 2 (15.2ml, 152mmol,30% in water) and the internal temperature is kept below 10 ℃. The mixture was heated at 60 ℃ under N 2 Stirring was continued for 1 hour. The mixture was cooled again to 30 ℃ and water (100 mL) and EtOH (100 mL) were added to the solution. A suspension was obtained which was filtered and concentrated in vacuo to give S-200-N19-4_4 (5 g, crude) as a solid.
1 H NMR(400MHz,CDCl3)δ5.44-5.32(m,1H),3.68-3.59(m,1H),3.39-3.35(m,1H),2.29-2.19(m,1H),2.08-1.89(m,4H),1.88-1.75(m,3H),1.62-1.60(m,2H),1.56-1.39(m,6H),1.36-1.24(m,3H),1.23-1.11(m,4H),1.08-0.98(m,4H),0.92-0.75(m,5H),0.70(s,3H)。
8. Dess-Martin periodinane (2.44g, 5.76mmol) was added to a solution of S-200-N19-4 \ (1g, 2.88mmol) in DCM (150 mL) at 25 ℃. The reaction was stirred at 25 ℃ for 1 hour. The reaction was stirred for 30mins at 25 ℃. The mixture was poured into saturated Na at 0 deg.C 2 S 2 O 3 (100 ml) and extracted with DCM (3 × 100 ml). The combined organic layers were washed with saturated NaHCO 3 (100mL. Times.2), brine (100 mL), na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product, which was purified by silica gel column (PE/EtOAc = 10) to give S-500-15-2 \u1 (800mg, 80%) as a solid.
1 H NMR(400MHz,CDCl3)δ9.58-9.57(m,1H),5.40-5.38(m,1H),2.37-2.35(m,1H),2.25-2.23(m,1H),2.08-1.76(m,7H),1.65-1.63(m,2H),1.53-1.37(m,5H),1.31-1.21(m,4H),1.19-1.00(m,6H),0.90-0.80(m,5H),0.73(s,3H)。
9. A solution of 1-bromo-3-methylbutane (4g, 26.4mmol) in THF (27 mL) was added dropwise to Mg (947mg, 39.5mmol) and I at 60 deg.C 2 (33.5mg, 0.132mmol) in THF (3 mL). The mixture was stirred at 60 ℃ for 1 hour. At N 2 Freshly prepared isopentyl magnesium bromide (30mL, 0.88M in THF, 26.4 mmol) was added to a solution of S-500-15-2-1 (800mg, 2.32mmol) in THF (2 mL) at 0 deg.C. The mixture was stirred at 0 ℃ for 1 hour. Adding NH to the mixture 4 Cl (50 mL, saturated aqueous solution). The mixture was extracted with EtOAc (2 × 50 mL). The combined organic phases were washed with brine (100 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product, which was purified by silica gel chromatography (PE/EtOAc =10/1 to 5/1) to give 44 (720mg, 75%) as a solid.
1 H NMR(400MHz,CDCl3)δ5.40-5.38(m,1H),3.63-3.61(m,1H),2.23-2.21(m,1H),2.10-1.74(m,7H),1.69-1.58(m,2H),1.54-1.34(m,8H),1.33-1.00(m,11H),0.95-0.75(m,14H),0.70(s,3H)。
LCMS Rt =1.289 min, 2 min chromatography, 30-90AB, 100% purity, MS ESI C 28 H 45 [M+H-2H 2 O]Calculated value 381 of + and found value 381.
10a at 25 ℃ in N 2 Next, to a solution of 44 (300mg, 0.720mmol) in THF (14 mL), benzoic acid (348mg, 2.85mmol) and triphenylphosphine (1.11g, 4.27mmol) were added. After stirring at 25 ℃ for 20mins, at 0 ℃ under N 2 Next, DIAD (780 mg, 3.86mmol) was added. The mixture was stirred at 0 ℃ for 20mins, then warmed to 25 ℃ and stirred at 25 ℃ for 17 hours. Water (100 mL) was added and the mixture was extracted with EtOAc (2 × 100 mL). The organic phase was washed with brine (100 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product to be purified (1.5 g, crude material).
10b at 25 ℃ in N 2 To a solution of 44 (1.9g, 4.55mmol) in THF (70 mL) was added benzoic acid (2.19g, 18.0 mmol) and triphenylphosphine (7.07g, 27.0 mmol) next. After stirring at 25 ℃ for 20mins, at 0 ℃ under N 2 Next, DIAD (4.93g, 24.4 mmol) was added. The mixture was stirred at 0 ℃ for 20mins, then warmed to 25 ℃ and stirred at 25 ℃ for 17 hours. Water (250 mL) was added and the mixture was extracted with EtOAc (2 × 250 mL). The organic phase was washed with brine (2X 300 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product. Combined with another batch from 300mg 44 to give the crude product, which was purified by silica gel column (PE/EtOAc = 8/1) to give S-500-15-1_1 (1.2 g, impure) as an oil, which was used directly in the next step.
11. To a solution of S-500-15-1 (1.2 g, impure) in THF/MeOH (2 mL/2 mL) at 25 deg.C was added NaOH (400 mg) and H 2 O (2 mL). The reaction was stirred at 50 ℃ for 16h. After cooling, the reaction mixture is washed with H 2 O (20 mL) diluted and extracted with EtOAc (2 × 30 mL). The combined organic layers were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo. The crude product was purified by silica gel column (PE/EtOAc = 4/1) to give product 3 (150 mg, impure), which was purified by trituration with MeCN (5 mL) at 25 ℃ to give 3 (30 mg, pure and 100mg, impure)Of (d) which is a solid.
1 H NMR(400MHz,CDCl3)δ5.39-5.37(m,1H),3.63-3.59(m,1H),2.26-2.21(m,1H),2.09-1.88(m,4H),1.86-1.76(m,2H),1.75-1.61(m,3H),1.54-1.32(m,7H),1.32-1.08(m,10H),1.07-0.96(m,1H),0.95-0.74(m,14H),0.95-0.74(m,1H),0.70(s,3H)。
LCMS Rt =1.281 min, chromatography over 2 min, 30-90AB, 98% purity, MS ESI C 28 H 47 O[M+H-H 2 O]Calculated value of + 399, found value 399.
Example 4: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -4- (4, 4-dimethylcyclohexyl) -3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (4)
Figure BDA0003762336660000911
Figure BDA0003762336660000921
1. t-BuLi (90.7mL, 118mmol,1.3M in n-hexane, 3.0 eq) was added to a solution of chloro (methoxymethyl) triphenylphosphorane (40.4g, 118mmol, 3.0eq) in THF (200 mL) at 0 ℃. After the addition, the reaction mixture was stirred at 0 ℃ for 1 hour. The mixture was added to a solution of S-500-6-29_2A (5g, 39.6mmol,1.0 eq) in THF (50 mL) at 0 ℃ and the reaction mixture was stirred at 15 ℃ for 2h. NH for the mixture 4 Cl (100ml, 10%) and extracted with EtOAc (2 × 200 mL). The organic phase was separated and concentrated in vacuo to give S-500-6-29_2B (18.0 g, crude material).
1 H NMR(400MHz,CDCl 3 )δ5.74(s,1H),3.52(s,3H),2.20-2.15(m,2H),1.95-1.90(m,2H),1.26-1.16(m,4H),0.90(s,6H)。
2. TFA (21.4 mL, 290mmol) was added to a stirred solution of S-500-6-29_2B (5.6 g, impure) in DCM (25 mL) at 15 ℃ and stirred for 1.5h at 15 ℃. The reaction mixture is usedAnd NaHCO 3 Aqueous (10 mL) quenched and extracted with EtOAc (2 × 20 mL). The combined organic layers were washed with anhydrous Na 2 SO 4 Dried and concentrated under reduced pressure to give S-500-6-29_2c (5.0 g, crude) as an oil, which was used in the next step without purification.
1 H NMR(400MHz,CDCl 3 )δ9.64(s,1H),2.15-2.05(m,1H),1.80-1.60(m,2H),1.70-1.35(m,4H),1.25-1.15(m,2H),0.91(s,3H),0.87(s,3H)。
3. At 15 ℃ in N 2 NaBH is reacted with 4 (1.61g, 42.7 mmol) was added to a solution of S-500-6-29-u 2C (5.0g, 35.6 mmol) in MeOH (50 mL). The mixture was stirred at 15 ℃ for 1 hour. The mixture was poured into water (50 mL) and stirred for 20 minutes. The aqueous phase was extracted with EtOAc (3 × 50 mL). The combined organic phases were washed with saturated brine (2 × 50 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated to give S-500-6-29 uk 2d (5.6 g, crude) as an oil.
1 H NMR(400MHz,CDCl 3 )δ3.47-3.42(m,2H),1.60-1.50(m,2H),1.42-1.30(m,4H),1.25-1.0(m,4H),0.91(s,3H),0.87(s,3H)。
4. At 15 ℃ in N 2 Next, tsCl (8.23g, 43.2mmol) was added to a solution of S-500-6-29-u 2D (5.6 g, 39.3mmol) in pyridine (50 mL). The mixture was stirred at 15 ℃ for 16 hours. The mixture was poured into water (50 mL) and stirred for 20 minutes. The aqueous phase was extracted with DCM (3 × 40 mL). The combined organic phases were washed with saturated brine (2X 200 mL), HCl (0.5M, 50ml), anhydrous Na 2 SO 4 Dried, filtered and concentrated to give an oil, which was recrystallized from hexane (50 mL) at 68 ℃ to give S-500-6-29_2e (4.2g, 61%) } as a solid.
1 H NMR(400MHz,CDCl 3 )δ7.80-7.76(m,2H),7.35-7.25(m,2H),3.86-3.80(m,2H),2.45(s,3H),1.60-1.45(m,3H),1.40-1.30(m,2H),1.20-1.05(m,4H),0.88(s,3H),0.82(s,3H)。
5. LiBr (2.33g, 26.9mmol) was added to a solution of S-500-6-29_2E (2g, 6.74mmol) in acetone (50 mL). Mixing the mixture inStirred at 65 ℃ for 12 hours. The mixture was quenched with water (50 mL) and extracted with MTBE (3 × 20 mL). The combined organic phases were washed with brine (50 mL) and Na 2 SO 4 Dried, filtered and concentrated to give S-500-6-29-2 (1.3 g, crude) as a liquid. Combined with another batch derived from 2.2g of S-500-6-29_2e, the combined crude product was filtered through a small polysiloxane gel and washed with PE (100 mL) and concentrated to give S-500-6-29_2 (2.6 g, 90%) as an oil.
1 H NMR(400MHz,CDCl 3 )δ3.34-3.28(m,2H),1.72-1.64(m,2H),1.60-1.48(m,1H),1.42-1.35(m,2H),1.28-1.18(m,4H),0.91(s,3H),0.87(s,3H)。
6. At 25 ℃ to Ph 3 To a solution of PMeBr (167g, 470mmol) in THF (900 mL) was added t-BuOK (52.7g, 470mmol). The reaction mixture was heated to 60 ℃ and stirred for 1 hour. Pregnenolone (50g, 157mmol) was added. The reaction mixture was stirred at 60 ℃ for 1 hour. Addition of saturated NH 4 Cl (900 mL). The mixture was extracted with EtOAc (2 × 1000 mL). The combined organic layers were washed with brine (2X 2000 mL) and Na 2 SO 4 Dried and concentrated in vacuo to give the crude product as an oil, which was purified by column chromatography on silica gel (PE: etOAc = 20.
1 H NMR(400MHz,CDCl 3 )δ5.40-5.30(m,1H),4.85(s,1H),4.71(s,1H),3.60-3.40(m,1H),2.40-2.20(m,2H),2.05-1.90(m,2H),1.85-1.60(m,9H),1.53-1.40(m,5H),1.25-0.90(m,9H),0.59(s,3H)。
7. To a solution of S-200-INT _1 (45g, 143mmol) in DCM (1500 mL) at 20 ℃ DMP (108g, 257 mmol) was added. The mixture was stirred at 20 ℃ for 2 hours. Water (800 mL) was added and NaHCO was added 3 (200 g of solid). The mixture was filtered. The filtrate was saturated with Na 2 S 2 O 3 (2X 2000 mL) washing with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solution of S-200-INT _2 in DCM (100 mL) which was used directly in the next step.
8. BHT (191g, 866mmol) was added to the formazan at 10 deg.CAlMe was added to a solution of benzene (500 mL) 3 (2M in toluene, 216mL, 433mmol) and stirring for 1 h. To this mixture was added a solution of S-200-INT _2 (theoretical mass: 44.6 g) in DCM (100 mL) at-78 ℃. The mixture was stirred at-78 ℃ for 1 hour. EtMgBr (141mL, 426 mmol) was added at-78 ℃. The mixture was stirred at-78 ℃ for 20mins. Saturated citric acid (1L) was added. The organic phase was separated, washed with brine (600 mL), and Na 2 SO 4 Dried and concentrated in vacuo to give the crude product, which was purified by column chromatography on silica gel (PE: etOAc =50 1 to 30.
1 H NMR(400MHz,CDCl 3 )δ5.35-5.25(m,1H),4.85(s,1H),4.71(s,1H),2.40-2.30(m,1H),2.10-1.60(m,14H),1.50-0.75(m,17H),0.58(s,3H)。
9. 9-BBN dimer (17.6g, 72.5 mmol) was added to a solution of S-200-INT-3E (5 g,14.5 mmol) in THF (40 mL). The mixture was heated at 60 ℃ under N 2 Stirring was continued for 3 hours to form a solid. Ethanol (8.33mL, 145mmol) and NaOH (28.9mL, 5M, 145mmol) were added to the reaction mixture. The mixture became clear. H is added dropwise at 25 DEG C 2 O 2 (14.4mL, 10M, 145mmol) and the internal temperature was raised to reflux (75 ℃). After addition the mixture was cooled and stirred for 1 hour to form a solid. To the mixture was added Na at 25 deg.C 2 SO 3 (20mL, 20. Degree. Aq.). The mixture was extracted with EtOAc (2 × 100 mL). The combined organic phases were washed with brine (2X 200 mL) and Na 2 SO 4 Dried, concentrated in vacuo, and purified by silica gel column (PE/EtOAc =10/1 to 3/1) to provide S-200-INT — 4E (3.5g, 67%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.68-3.60(m,1H),3.41-3.32(m,1H),2.40-2.32(m,1H),2.03-1.93(m,2H),1.92-1.65(m,4H),1.58-1.16(m,13H),1.16-0.90(m,11H),0.90-0.81(m,3H),0.73-0.62(s,3H)。
10. DMP (4.66g, 11.0 mmol) was added to a solution of S-200-INT _4E (2g, 5.54mmol) in DCM (30 mL) at 25 ℃. The reaction mixture was stirred at 25 ℃ for 10 minutes. Reaction mixtureThe mixture was quenched with saturated NaHCO at 25 deg.C 3 Aqueous solution (30 mL) was quenched. The DCM layer was separated and the aqueous phase was extracted with DCM (30 mL). The combined organic phases were saturated with Na 2 SO 3 Aqueous (3X 50 mL), brine (50 mL), na 2 SO 4 Dried, filtered and concentrated in vacuo to give S-200-INT _5E (2.0 g, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.59-9.56(m,1H),5.31-5.26(m,1H),2.42-2.10(m,2H),2.10-1.80(m,4H),1.79-1.54(m,7H),1.54-1.31(m,7H),1.28-0.90(m,9H),0.90-0.81(m,4H),0.73(s,3H)。
11. A solution of S-500-6-29 (2.56g, 12.5 mmol) in THF (8 mL) was added dropwise to Mg (600mg, 25.0 mmol) and I at 75 deg.C 2 (63.4 mg, 0.25mmol) in THF (3 mL). The mixture was stirred at 75 ℃ for 1 hour. After cooling, a solution of S-500-6-1_1 (1g, 2.78mmol) in THF (30 mL) was added slowly at 15 ℃. After addition, the mixture was stirred at 15 ℃ for 2hrs with saturated NH 4 Cl (40 mL) and saturated citric acid (20 mL) were quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases were washed with brine (2 × 30 mL) and Na 2 SO 4 Dried, filtered and concentrated and purified by combi-flash (0-15% etoac in PE) to give S-500-6-29 \ u 1 mixture (800mg, 60%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.33-5.19(m,1H),3.88-3.71(m,1H),2.42-2.29(m,1H),2.07-1.86(m,4H),1.78-1.59(m,4H),1.54-1.31(m,13H),1.29-1.13(m,8H),1.12-0.99(m,8H),0.94-0.79(m,13H),0.68(s,3H)。
12. DMP (1.39g, 3.30mmol) was added to a solution of S-500-6-29-1 (800mg, 1.65mmol) in DCM (30 mL). Then, the reaction mixture was stirred at 15 ℃ for 10 minutes. The reaction mixture was saturated with NaHCO 3 The aqueous solution (50 mL) was quenched until the pH of the aqueous layer was about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (20 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3X 40 mL), saturated NaHCO 3 (40 mL), brine (40 mL), na 2 SO 4 Drying the mixtureFiltered and concentrated to give crude S-500-6-29 \u2 (800 mg, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.31-5.25(m,1H),2.54-2.43(m,1H),2.40-2.21(m,3H),2.07-1.87(m,3H),1.81-1.57(m,7H),1.53-1.39(m,7H),1.38-1.29(m,3H),1.27-1.16(m,4H),1.15-1.04(m,8H),1.03(s,3H),1.00-0.92(m,2H),0.91-0.80(m,9H),0.69(s,3H)。
13. NaBH was added 5 times every 5 minutes 4 (2.80g, 82.5 mmol) was added to a solution of S-500-6-29 (800mg, 1.65mmol) in MeOH (5 mL) and THF (5 mL). The mixture was stirred at 15 ℃ for 30 minutes. The mixture is saturated with NH 4 Cl (50 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases are washed with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 49 (290mg, 36%) and 12 (120mg, 45%) as solids.
49:
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.85-3.77(m,1H),2.40-2.32(m,1H),2.07-1.87(m,4H),1.76-1.69(m,1H),1.66-1.55(m,5H),1.53-1.42(m,7H),1.41-1.31(m,5H),1.30-1.12(m,8H),1.11-1.05(m,3H),1.03(s,3H),1.01-0.92(m,2H),0.91-0.82(m,12H),0.68(s,3H)。
LCMS Rt =1.718 min, chromatography at 2.0 min, 30-90AB _E, 98% purity, MS ESI C 33 H 53 [M+H-2H 2 O] + Calculated value 449, found value 449.
12:
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.85-3.77(m,1H),2.40-2.32(m,1H),2.06-1.95(m,3H),1.77-1.58(m,7H),1.54-1.28(m,12H),1.27-1.06(m,11H),1.03(s,3H),1.00-0.95(m,2H),0.93-0.82(m,12H),0.69(s,3H)。
LCMS Rt =1.708 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 33 H 53 [M+H-2H 2 O] + Calculated 449, found 449.
14. Pd (OH) 2 (200 mg, anhydrous) was added to a solution of 49 (140mg, 0.288mmol) in MeOH (30 mL). The mixture was heated at 50 ℃ in H 2 Next (50 Psi) was stirred for 48 hours. The mixture was filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 59 (27mg, 19%) and 4 (42mg, 30%) as solids.
4:
1 H NMR(400MHz,CDCl 3 )δ3.84-3.76(m,1H),1.98-1.85(m,2H),1.69-1.54(m,9H),1.53-1.46(m,3H),1.45-1.28(m,9H),1.27-1.20(m,4H),1.19-1.13(m,5H),1.12-1.02(m,4H),1.01-0.92(m,2H),0.91-0.85(m,12H),0.82(s,3H),0.70-0.61(m,4H)。
LCMS Rt =1.799 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 33 H 55 [M+H-H 2 O] + Calculated value 451 of (d), found value 451.
Example 5: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-6, 6-dimethylhept-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (5)
Figure BDA0003762336660000971
1. At N 2 A solution of 1-bromo-3, 3-dimethylbutane (3.68g, 22.3mmol) in THF (8 mL) was added dropwise to Mg (1.08g, 44.6 mmol) and I at 50-55 deg.C 2 (1 mg) in THF (2 mL). The mixture was stirred at 55 ℃ for 1 hour. A solution of S-500-6-1_1 (0.8g, 2.23mmol) in THF (5 mL) was then added to freshly prepared (3, 3-dimethylbutyl) magnesium bromide (22.3 mmol in 10mL THF) at 0 deg.C. The mixture was stirred at 15 ℃ for 2 hours. Citric acid (20mL, 10% aq.) was added to the mixture. The mixture was extracted with EtOAc (30 mL). The organic layer was separated and concentrated in vacuo to give a mixture which was subjected to flash column separation (0-15% etoac in PE) to give 5 (580 mg, p1, 58%) and 54 (50mg, 5% impure).
5:
1 H NMR(400MHz,CDCl 3 )δ5.33-5.24(m,1H),3.65-3.54(m,1H),2.41-2.31(m,1H),2.11-1.84(m,4H),1.76-1.38(m,15H),1.38-1.00(m,12H),0.93-0.80(m,15H),0.70(s,3H)。
54:
1 H NMR(400MHz,CDCl 3 )δ5.33-5.24(m,1H),3.62-3.52(m,1H),2.41-2.31(m,1H),2.11-1.90(m,3H),1.75-1.00(m,28H),1.00-0.75(m,18H),0.70(s,3H)。
2. DMP (1.1g, 2.6 mmol) and water (1 drop) were added to a solution of 5 (580 mg,1.3 mmol) in DCM (10 mL) at 20 ℃. The mixture was stirred at 20 ℃ for 2h. Saturated NaHCO 3 Solution (20 mL) and Na 2 S 2 O 3 (20mL, sat.) was added to the mixture. The mixture was extracted with EtOAc (50 mL). Organic layer with NaHCO 3 /Na 2 S 2 O 3 (20+20mL, sat.) twice, washing with Na 2 SO 4 Drying, filtration, and concentration in vacuo afforded S-500-6-1_3 (520mg, 90%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.38-5.18(m,1H),2.62-2.22(m,4H),2.11-1.85(m,3H),1.78-1.57(m,7H),1.57-1.32(m,8H),1.32-1.21(m,2H),1.19-1.09(m,5H),1.08-1.01(m,4H),1.00-0.91(m,1H),0.90-0.80(m,12H),0.70(s,3H)。
3. NaBH is reacted at 15 ℃ 4 (1.77g, 46.8 mmol) was added portionwise to a solution of S-500-6-1_3 (520mg, 1.17mmol) in THF (5 mL) and MeOH (10 mL). The mixture was stirred at 15 ℃ for 20 minutes. The mixture is treated with NH 4 Cl (20 mL, saturated aqueous) was quenched and extracted with EtOAc (50 mL). The organic layer was separated and concentrated in vacuo to give a mixture which was separated by flash column (0-15% etoac in PE) to give 5 (300 mg, impure) and 54 (170 mg, impure).
4. Impure 5 (300 mg, impure) was purified by flash column (0-12% etoac in PE) to give a solid. The solid was dissolved in MeCN (50 mL) at 60 ℃ and concentrated in vacuo to give 5 (270mg, 52%) as a solid.
5:
1 H NMR(400MHz,CDCl 3 )δ5.33-5.24(m,1H),3.67-3.54(m,1H),2.41-2.31(m,1H),2.11-1.84(m,4H),1.78-1.57(m,5H),1.55-1.38(m,12H),1.38-1.07(m,7H),1.03(s,3H),0.93-0.89(m,12H),0.85(t,J=7.6Hz,3H),0.70(s,3H)。
LCMS Rt =5.587 min, chromatography at 7.0 min, 30-90_AB _E, purity 96.5%, MS ESI C 30 H 49 [M+H-2H 2 O] + The calculated value 409 of (1) is actually measured value 409.
Example 6: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -17- ((2S, 3R) -5-cyclopropyl-3-hydroxypentan-2-yl) -3-ethyl-10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (6)
Figure BDA0003762336660000991
1. At N 2 A solution of (2-bromoethyl) cyclopropane (1.8g, 12mmol) in THF (8 mL) was added dropwise to Mg (641mg, 26.4mmol) and I at 50-55 deg.C 2 (1 mg) in THF (2 mL). After stirring at 55 ℃ for 1h, the mixture was diluted with THF (10 mL). The Grignard solution was added to a solution of S-500-6-1_1 (0.8g, 2.23mmol) in THF (10 mL) at 0 ℃. After stirring for 4hrs at 15 deg.C, the reaction was quenched with NH 4 Cl (20mL, 10 aq.) was quenched and extracted with EtOAc (30 mL). The organic layer was separated and concentrated in vacuo to give a mixture (1 g, crude) as a solid which was separated by flash column (0-25% DCM/EtOAc (1/1) in PE) to give S-500-6-20 (700mg, 73%, impure), and S-500-6-19 (70mg, 7%, impure) as a solid.
2. DMP (1.38g, 3.26mmol) and water (1 drop) were added to a solution of S-500-6-20 (700mg, 1.63mmol) in DCM (10 mL) at 20 ℃. After stirring for 2h at 20 ℃ the mixture was taken up in NaHCO 3 (20mL, sat.) and Na 2 S 2 O 3 (20mL, sat.) and extracted with EtOAc (50 mL). The organic layer was washed with saturated NaHCO 3 /Na 2 S 2 O 3 (2X (20 mL/20 mL)) and washed with Na 2 SO 4 Dried, filtered, and concentrated in vacuo to afford S-500-6-19_3 (700mg, 100%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.35-5.20(m,1H),2.72-2.26(m,4H),2.17-1.87(m,3H),1.82-1.35(m,13H),1.35-1.20(m,2H),1.20-0.91(m,12H),0.85(t,J=7.2Hz,3H),0.80-0.62(m,4H),0.53-0.33(m,2H),0.12-0.00(m,2H)。
3. NaBH is reacted at 15 ℃ 4 (2.46g, 65.1mmol) was added portionwise to a solution of S-500-6-1/-3 (700mg, 1.63mmol) in THF (5 mL) and MeOH (5 mL). After stirring for 20mins at 15 ℃ the mixture is taken up in NH 4 Cl (20 mL, saturated aqueous) was quenched and extracted with EtOAc (50 mL). The organic layer was separated and concentrated in vacuo to give 760mg of the mixture as a solid, which was separated by flash column (0-35% DCM/EtOAc (1/1) in PE) to give 69 (330mg, 47%) and 6 (250mg, 35%, impure) as solids. Impure 6 (250 mg) was further separated by flash column (0-35% DCM/EtOAc (1/1) in PE) to give 6 (170mg, 23%) as a solid.
6:
1 H NMR(400MHz,CDCl 3 )δ5.32-5.24(m,1H),3.77-3.66(m,1H),2.41-2.31(m,1H),2.09-1.91(m,3H),1.79-1.59(m,6H),1.55-1.21(m,14H),1.21-1.06(m,4H),1.03(s,3H),1.00-0.95(m,1H),0.93(d,J=6.8Hz,3H)0.85(t,J=7.6Hz,3H),0.70(s,3H),0.68-0.62(m,1H),0.49-0.38(m,2H),0.11-0.02(m,2H)。
LCMS Rt =1.380 min, chromatography at 2.0 min, 30-90_AB _E, 100% purity, MS ESI C 29 H 47 O[M+H-H 2 O] + The calculated value 411 of (a), the actual measurement value 411.
Example 7: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((1R, 2S) -1-hydroxy-1- (pyridin-3-yl) propan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (7)
Figure BDA0003762336660001011
1. In N 2 Trimethylaluminum (2M in toluene, 469mL, 939mmol) was added dropwise to a solution of BHT (416g, 1.88mol) in toluene (1500 mL) at 0 deg.C. The mixture was stirred at 0 ℃ for 30mins and used directly as a solution of MAD (0.47M in toluene) without further purification. At N 2 Next, a solution of N-005 (100g, 315mmol) in toluene (800 mL) was added dropwise to a solution of MAD (0.47M in toluene, 2.01L, 945mmol) at-70 ℃. The mixture was stirred at-70 ℃ for 30mins. EtMgBr (3M in ether, 315mL, 945mmol) was added dropwise to the above mixture. The resulting mixture was stirred at-70 ℃ for 1 hour. The reaction mixture was poured into ice-cooled aqueous citric acid (1000 mL) and extracted with EtOAc (2 × 600 mL). The combined organic layers were washed with brine (500 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (0-20% etoac in PE) to give 85g of N-005 _2as a solid (78% yield).
1 H NMR(400MHz,CDCl 3 )δ2.55-2.46(m,1H),2.19-2.12(m,1H),2.11-2.09(m,3H),2.08-1.96(m,1H),1.71-1.48(m,10H),1.47-1.31(m,5H),1.30-1.09(m,7H),1.06-0.94(m,2H),0.92-0.87(m,3H),0.86-0.79(m,3H),0.75-0.64(m,1H),0.60(s,3H)。
2. At 15 ℃ in N 2 Downward MePPh 3 To a suspension of Br (174g, 0.49mol) in THF (1000 mL) was added t-BuOK (54.9 g, 0.49mol). After stirring at 50 ℃ for 30mins, a solution of N-005_2 (85g, 2450 mmol) in THF (800 mL) was added portionwise at below 65 ℃. The mixture was stirred at 50 ℃ for 1 hour with NH 4 Quenched with Cl (1000 mL) and extracted with EtOAc (2X 900 mL). The organic layer was separated and concentrated in vacuo to give the crude product, which was triturated from MeOH/water (1.5l, 1. After cooling, the mixture was filtered and the filter cake was washed with MeOH/water (2x500ml, 1), concentrated in vacuo to afford N-005 (75 g, crude) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.85-4.82(m,1H),4.71-4.68(m,1H),2.06-1.94(m,1H),1.86-1.78(m,1H),1.76-1.71(m,4H),1.70-1.62(m,4H),1.61-1.48(m,6H),1.47-1.30(m,3H),1.29-1.05(m,8H),1.04-0.92(m,1H),0.91-0.82(m,6H),0.76-0.63(m,1H),0.56(s,3H)。
3. In N 2 To a solution of N-005 (75g, 217mmol) in THF (1800 mL) was added 9-BBN dimer (105g, 434mmol) below. The mixture was stirred at 60 ℃ for 3 hours. Ethanol (124ml, 2.17mol) and aqueous NaOH (434ml, 5m, 2.17mmol) were added portionwise to the reaction mixture. Then H is added dropwise at 0 DEG C 2 O 2 (217mL, 10M, 2.17mol). The mixture was warmed to 65 ℃ and stirred for 1hr and diluted with water (1.5L). The reaction mixture was extracted with EtOAc (2 × 800 mL). To the combined organic layers was added saturated Na 2 S 2 O 3 Aqueous solution (600 mL) and stirred for 1 hour. The reaction was checked by potassium iodide-starch paper to confirm excess H 2 O 2 Is destroyed. The organic phase was then washed with saturated brine (2X 500 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-005 (78 g, crude) as a solid. Crude N-005 \u4 (78 g, impure) was triturated from MeOH/H2O =10/1 at 15 ℃ to give N-005 \u4 (70 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.68-3.60(m,1H),3.41-3.32(m,1H),1.99-1.92(m,1H),1.88-1.75(m,1H),1.69-1.45(m,10H),1.44-1.29(m,4H),1.28-1.15(m,6H),1.14-0.91(m,8H),0.90-0.79(m,7H),0.67(s,3H)。
4. To a solution of N-005 (70g, 193mmol) in DCM (800 mL) was added DMP (122g, 289mmol). The reaction was then stirred for 30mins at 15 ℃. Adding saturated NaHCO to the reaction mixture 3 Aqueous (500 mL) solution and stirred at 15 ℃ for 20mins. Adding saturated Na 2 S 2 O 3 Aqueous solution (600 mL) and the mixture was stirred at 15 ℃ for an additional 1 hour. The reaction was checked by potassium iodide-starch paper to confirm that excess DMP was destroyed. The aqueous phase was extracted with DCM (2 × 400 mL). The combined organic layers were washed with saturated NaHCO 3 (400 mL) aqueous solution and brine (400 mL) and washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-005 (70 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.58-9.55(m,1H),2.39-2.30(m,1H),1.95-1.78(m,2H),1.69-1.42(m,10H),1.41-1.30(m,4H),1.29-1.14(m,5H),1.13-0.95(m,6H),0.94-0.86(m,4H),0.85-0.81(m,3H),0.69(m,4H)。
5. i-PrMgCl (2.49mL, 4.98mmol,2M in ether) was added dropwise to a solution of 3-bromopyridine (875mg, 5.54mmol) in THF (5 mL). After stirring for 1h at 25 deg.C, a solution of N-8-7_1 (200mg, 0.554mmol) in THF (5 mL) was added. After stirring for 16hrs at 25 deg.C, the reaction mixture was washed with NH 4 Cl (50ml, 10% aq.) and extracted with EtOAc (2 × 20 mL). The combined organic phases are washed with Na 2 SO 4 Dried, filtered, concentrated and purified by flash column (0-50% EtOAc in DCM) to give N-8-19 \u1 (100mg, 41%) as a solid.
6.N-8-19 \, 1 (100mg, 0.227mmol) was separated by SFC (column: AD (250mm 30mm, 5um), gradient: 50-50% B (A =0.05% 3 /H 2 O, B = MeOH), flow rate: 80 mL/min) to give 7 (Peak 1, 57mg, 57%) and 89 (Peak 2,8mg, 8%) as solids.
SFC peak 1: rt =1.798 min and peak 2Rt = -1.985 min, AD-H _3UM _4_5_40_4ML ("Chiralpak AD-3 50 × 4.6mm I.D.,3um mobile phase: A: CO: 1.985 min, chromatography 3 min 2 B: isopropanol (0.05% DEA) gradient: 5 to 40% in 1.4 minutes and held at 40% for 1.05 minutes, then 5% b for 0.35 minutes, flow rate: 4mL/min, column temperature: 40 deg.C ").
7:
1 H NMR(400MHz,CDCl 3 )δ8.56-8.52(m,1H),8.49-8.45(m,1H),7.68-7.62(m,1H),7.29-7.24(m,1H),5.01-4.95(m,1H),2.11-2.01(m,1H),1.96-1.89(m,1H),1.83-1.76(m,1H),1.73-1.63(m,4H),1.59-1.47(m,6H),1.43-1.29(m,4H),1.27-1.20(m,4H),1.19-1.06(m,4H),1.03-0.92(m,1H),0.91-0.85(m,4H),0.83(s,3H),0.77-0.73(m,3H),0.70-0.64(m,4H)。
LCMS Rt =1.017 min, chromatography at 2.0 min, 10-80AB _E, purity 100%, MS ESI C 29 H 46 NO 2 [M+H] + Is calculated by440, found value 440.
SFC Rt =1.780 min, AD-H _3UM _4_5_40_4ML,100% by 3 min chromatography.
Example 8: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -3,10, 13-trimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (8)
Figure BDA0003762336660001041
1. At a temperature below 25 ℃ in N 2 AlMe was added dropwise to a solution of BHT (1.97kg, 8.94mol) in toluene (1L) under an atmosphere 3 (2.14L, 2.0M in toluene, 4.28 mol). The resulting mixture was stirred at 25 ℃ for 1 hour. A solution of S-200-INT _2 (794 g,85% wt, 2.16 mol) in DCM (3L) was added at-70 ℃. The mixture was stirred at-70 ℃ for 1 hour. MeMgBr (862mL, 3.0M in ether, 2.59 mol) was added at-70 ℃. The reaction mixture was stirred at-70 ℃ for 10mins. The mixture was quenched by saturated citric acid (3L) and extracted with EtOAc (2 x 2L). The combined organic phases were washed with brine (2L) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a residue, which was triturated from MeCN (3L) at 25 ℃ to give S-200-INT _3 (340g, 43%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.34-5.26(m,1H),4.85(s,1H),4.71(s,1H),2.50-2.35(m,1H),2.07-1.94(m,3H),1.91-1.84(m,1H),1.83-1.63(m,8H),1.58-1.33(m,6H),1.27-1.13(m,3H),1.12(s,3H),1.10-1.05(m,1H),1.02(s,3H),1.00-0.92(m,1H),0.58(s,3H)。
2. At 15 ℃ in N 2 THF (1L) was added to a mixture of S-200-INT _3 (149g, 453 mmol) and 9-BBN dimer (127g, 520mmol). The reaction mixture was stirred at 60 ℃ for 1 hour. The mixture was cooled to 15 ℃. EtOH (208g, 4.53mol) was added at 15 ℃. Aqueous NaOH (906 mL,5M, 4.53mol) was added dropwise at 15 ℃. H is added dropwise at 15 DEG C 2 O 2 (514g, 30%,4.53 mol). The resulting mixture was stirred at 60 ℃ for 1 hour. A solid is produced. The solid was washed with ethanol (200 mL) To give a solid, which was triturated continuously with EtOH (2.3L) at reflux and water (2.5L) at 80 ℃ to give 15-3b (131g, 84%) as a solid. The ethanol-derived filtrate was concentrated in vacuo to give 15-3b (30 g, crude) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.35-5.24(m,1H),3.67-3.61(m,1H),3.42-3.33(m,1H),2.50-2.35(m,1H),2.07-1.92(m,3H),1.88-1.65(m,3H),1.60-1.38(m,9H),1.37-1.26(m,1H),1.26-1.12(m,4H),1.11(s,3H),1.08(s,1H),1.05(d,J=6.8Hz,3H),1.01(s,3H),1.00-0.91(m,1H),0.70(s,3H)。
3. DMP (2.44g, 5.76mmol) was added to a solution of 15-3b (1g, 2.88mmol) in DCM (10 mL). The reaction was then stirred at 25 ℃ for 10 minutes. The reaction mixture was purified by addition of saturated NaHCO 3 Aqueous solution (20 mL) and saturated Na 2 S 2 O 3 Quench (20 mL) with water and extract with DCM (2X 50 mL). The combined organic layers were washed with saturated NaHCO 3 Aqueous solution (3X 50 mL) and brine (50 mL) were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give S-500-2-9_1 (1 g, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.57(brs,1H),5.35-5.25(m,1H),2.50-2.30(m,2H),2.05-1.95(m,3H),1.95-1.80(m,1H),1.75-1.65(m,1H),1.65-1.60(m,3H),1.55-1.50(m,2H),1.50-1.40(m,2H),1.40-1.30(m,1H),1.25-1.20(m,2H),1.20-1.15(m,2H),1.15-1.10(m,6H),1.05-0.95(m,5H),0.90-0.70(m,1H),0.68(s,3H)。
4. Magnesium (641mg, 26.4mmol) and I 2 (33.5mg, 0.132mmol) at 60 ℃ with stirring and N 2 A solution of isoamyl magnesium bromide (2g, 13.2mmol) in THF (20 mL) was added dropwise. Then, the reaction mixture was stirred at 60 ℃ for 1 hour. The reaction mixture was used directly as the isopentyl magnesium bromide solution without any purification. At 0 ℃ in N 2 Next, the Grignard solution was added to a solution of S-500-2-9-u 1 (1g, 2.90mmol) in THF (10 mL). Then, the reaction mixture was stirred at 25 ℃ for 1 hour. Adding saturated NH to the reaction mixture 4 Aqueous Cl (50 mL), extracted with EtOAc (2X 50 mL), washed with brine (50 mL), na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product. The crude product was purified by silica gel column (EtOAc/PE = 1/4) to give impure S-500-2-9-1A (560 mg) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.28-5.25(m,1H),3.90-3.80(m,0.25H),3.68-3.58(m,0.75H),2.48-2.36(m,1H),2.05-1.95(m,3H),1.95-1.80(m,1H),1.80-1.75(m,1H),1.75-1.52(m,6H)1.52-1.42(m,6H),1.42-1.32(m,3H),1.32-1.22(m,3H),1.22-1.12(m,3H),1.12-1.02(m,2H),1.01(s,3H),1.00-0.92(m,1H),0.92-0.85(m,9H),0.85-0.77(m,1H),0.69(s,3H)。
S-500-2-9-1A (560 mg) was purified by SFC (column: chiralcel OD-3X 4.6mm I.D.,3um mobile phase: A: CO 2B: ethanol (0.05% DEA) gradient: 5% to 40% B in 5 minutes and held at 40% for 2.5 minutes, then 5% B was held for 2.5 minutes at a flow rate: 2.5mL/min, column temperature: 35 ℃) to obtain impure solid 30 (160 mg) and solid 75 (265mg, 47%).
1 H NMR(400MHz,CDCl 3 )δ5.35-5.30(m,1H),3.70-3.60(m,1H),2.50-2.40(m,1H),2.05-1.90(m,4H),1.85-1.75(m,2H),1.75-1.60(m,1H),1.55-1.45(m,8H),1.45-1.25(m,8H),1.25-1.10(m,4H),1.10-1.05(m,2H),1.02(s,3H),0.99-0.91(m,3H),0.91-0.89(m,4H),0.88(s,3H),0.69(s,3H)。
LCMS Rt =1.162 min, chromatography over 1.5 min, 5-95AB, 99% purity, MS ESI C 28 H 45 [M+H-2H 2 O] + Calculated value 381, found value 381 of (1).
6. Will be anhydrous Pd (OH) 2 the/C (100 mg) was added to a solution of 75 (230mg, 0.551mmol) in THF (5 mL) and MeOH (5 mL). At H 2 And the reaction mixture was stirred at 50 ℃ for 24h at 50 Psi. HNMR then showed the reaction was complete. The reaction mixture was filtered through filter paper and concentrated in vacuo to give an impure product. The impure product was recrystallized from MeCN (3 mL) to give 8 (68mg, 30%) as an off-white solid.
1 H NMR(400MHz,CDCl 3 )δ3.65-3.55(m,1H),2.00-1.80(m,2H),1.76-1.60(m,3H),1.55-1.48(m,3H),1.48-1.38(m,4H),1.38-1.26(m,7H),1.26-1.23(m,4H),1.23-1.06(m,5H),1.06-1.02(m,3H),1.02-095(m,1H),0.95-0.85(m,10H),0.81(s,3H),0.70-0.60(m,4H)。
LCMS Rt =1.171 min, chromatography at 1.5 min, 5-95AB, purity 100%.
MS MS ESI C 28 H 47 [M+H-2H 2 O] + Calculated value 383 of (d), found value 383.
Example 9: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3- (methoxymethyl) -10, 13-dimethyl-17- ((2S, 3R) -4, 4-trifluoro-3-hydroxybut-2-yl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (9)
Figure BDA0003762336660001071
Figure BDA0003762336660001081
1. Pd/C (5 g, <1% water) was added to a solution of N-004-022/1 (50g, 151mmol) in MeOH (100 mL) and THF (100 mL). The solution was then hydrogenated at 30psi hydrogen at 25 ℃ for 48 hours. The mixture was filtered through a pad of celite and the filtrate was concentrated in vacuo to give N-004-022_2 (50 g, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.65-3.55(m,1H),3.40-3.3(m,1H),2.80-2.60(m,1H),2.40-2.30(m,1H),2.25-2.10(m,1H),2.10-1.95(m,3H),1.80-1.65(m,3H),1.65-1.53(m,1H),1.53-1.40(m,4H),1.40-1.01(m,17H),0.70(s,3H)。
2. In N 2 A stirred solution of trimethyliodosulfide (19.8g, 90.2mmol) and t-BuOK (10.1g, 90.2mmol) in DMSO (200 mL) was heated at 60 ℃ for 1hr. N-004-022 u 2 (15g, 45.1mmol) was added to the reaction mixture and stirred at 60 ℃ for 10mins. For reactionsWater (1000 mL) and extracted with EtOAc (2 × 500 mL). The combined organic phases were washed with water (2X 500 mL), brine (300 mL), and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-004-022_3 (15.5 g, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.18-4.08(m,1H),3.67-3.60(m,1H),3.40-3.30(m,1H),2.70-2.50(m,3H),2.40-2.30(m,1H),2.01-1.50(m,14H),1.40-0.65(m,14H),0.68(s,3H)。
3. At 25 ℃ in N 2 MeONa (12.0g, 223mmol) was then added to a solution of N-004-022 \3 (15.5g, 44.7mmol) in MeOH (500 mL). Mixing the mixture in N 2 The mixture was stirred at 70 ℃ under reflux for 16hrs. The reaction was treated with water (500 mL). The aqueous phase was extracted with DCM (2 × 300 mL). The combined organic phases were washed with saturated brine (2X 300 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated to give N-004-022 \4 (15 g, crude material) as a solid. Crude N-004-022 \u4 (15 g) was purified by silica gel chromatography (PE/EtOAc =10/1 to 5/1) to give N-004-022 \u4 (7.4g, 50%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.76-3.73(m,1H),3.64-3.60(m,1H),3.40-3.33(m,4H),3.22-3.16(m,2H),2.01-1.69(m,6H),1.62-1.51(m,4H),1.44-1.31(m,13H),1.10-0.99(m,5H),0.97(s,3H),0.67(s,3H)。
4. DMP (1.56g, 3.69mmol) was added to a solution of N-004-022_4 (1.4 g, 3.69mmol) in DCM (15 mL). Then, the reaction mixture was stirred at 25 ℃ for 10 minutes. The reaction mixture was saturated with NaHCO 3 Aqueous solution (20 mL) was quenched until pH =9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (20 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3 × 10 mL), saturated NaHCO 3 The solution (10 mL), brine (20 mL) was washed with Na 2 SO 4 Dried, filtered and concentrated. The residue was triturated with MeCN (10 mL) to give N-004-022 u 5 (700, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.56-9.58(m,1H),3.39(s,3H),3.24-3.18(m,2H),2.40-2.31(m,1H),2.01-1.50(m,11H),1.47-1.01(m,16H),0.97(s,3H),0.70(s,3H)。
Figure BDA0003762336660001091
5. At N 2 TMSCF was added to a solution of N-004-022_5 (200mg, 0.531mmol), csF (40.2mg, 0.265mmol) in THF (5 mL) at 0 deg.C 3 (187mg, 1.32mmol). The mixture was stirred at 25 ℃ for 1 hour. To the mixture was added TBAF.3H2O (836 mg, 2.65mmol). After stirring for 2hrs at 25 ℃ the mixture was 50% NH 4 Cl (20 mL) quenched and extracted with EtOAc (2 × 10 mL). The combined organic phases were washed with brine (20 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, PE/EA = 10/1) to give 9 (56mg, 24%) and 71 (30 mg, impure) as white solids.
9:
1 H NMR(400MHz,CDCl 3 )δ4.05-3.95(m,1H),3.39(s,3H),3.24-3.18(m,2H),2.00-1.83(m,5H),1.77-1.68(m,2H),1.64-1.47(m,8H),1.43-1.35(m,5H),1.31-1.08(m,6H),1.06-1.00(m,3H),0.97(s,3H),0.70(s,3H)。
LCMS Rt =1.156 min, chromatography over 2min, 30-90AB (u 2min. Lcm) purity 100%, MS ESI C 25 H 41 F 3 O 3 [M+Na] + Calculated value of 469, found 469.
Example 10: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((1S, 2S) -1-cyclopropyl-1-hydroxypropan-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (10)
Figure BDA0003762336660001101
Figure BDA0003762336660001111
1. A solution of N-8-7-u 1 (500mg, 1.38mmol) in THF (5 mL) was added to a solution of cyclopropyl magnesium bromide (1g, 13.7mL,0.5M in THF) in THF (5 mL) at 0 deg.C and stirred at 25 deg.C for 4hrs. Adding NH to the mixture 4 Cl (20mL, 10% aq.) and extracted with EtOAc (2X 30 mL). The organic layer was separated and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography eluting with PE/EtOAc =1/1 to give N-8-13_1 (140mg, 25%) as a solid.
LCMS Rt =1.192 min, chromatography at 2.0 min, 30-90AB _2MIN _E, purity 99%, MS ESI C 27 H 43 [M+H-2H 2 O] + The calculated value 367 and the actual value 367 of (a).
2. DMP (294mg, 0.694mmol) was added to a solution of N-8-13- \ u 1 (140mg, 0.347mmol) in DCM (5 mL). The reaction mixture was then stirred at 25 ℃ for 1h. The reaction mixture was saturated with NaHCO 3 The aqueous solution (50 mL) was quenched until the pH of the aqueous layer became about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (100 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3X 100 mL), saturated NaHCO 3 (100 mL), brine (40 mL), na 2 SO 4 Dried, filtered and concentrated to give N-8-13 \u2 (140 mg, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.65-2.55(m,1H),1.95-1.90(m,2H),1.50-1.15(m,19H),1.14-0.95(m,7H),0.94-0.80(m,12H),0.69(s,3H)。
3. Reacting NaBH 4 (1.18g, 17.4mmol) was added five times, every 5 minutes, to a solution of N-8-13_2 (140mg, 0.347mmol) in MeOH (1 mL) and THF (1 mL). The mixture was stirred at 15 ℃ for 30 minutes. The mixture is saturated with NH 4 Cl (50 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases are treated with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (25% etoac in PE) to give solid 10 (26mg, 19%) and solid 90 (12mg, 9%).
10:
1 H NMR(400MHz,CDCl 3 )δ2.85-2.80(m,1H),2.00-1.95(m,1H),1.90-1.80(m,1H),1.55-1.10(m,16H),1.09-0.80(m,17H),0.70-0.60(m,5H),0.58-0.43(m,3H),0.32-0.34(m,1H),0.13-0.06(m,1H)。
LCMS Rt =3.840 min, chromatography over 7.0 min, 30-90AB u 7MIN \ u E, 97% purity, MS ESI C 27 H 43 [M+H-2H 2 O] + The calculated value 367 and the actual value 367 of (a).
HPLC Rt =13.470 min, chromatography over 30min, 70-90ab \u1 _30min. M, purity 97%.
Example 11: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-10, 13-dimethyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybutan-2-yl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (11)
Figure BDA0003762336660001121
Figure BDA0003762336660001131
1. Subjecting TMSCF to a reaction at 0 deg.C 3 (493mg, 3.47mmol) was added to a solution of S-500-6-1_1 (500mg, 1.39mmol) and CsF (105mg, 695umol) in THF (5 mL). The mixture was stirred at 25 ℃ for 1hr and washed with TBAF.3H 2 O (1.09g, 3.47mmol). The mixture was stirred at 25 ℃ for 2hrs, quenched with water (100 mL) and extracted with EtOAc (2 × 50 mL). The combined organic phases were washed with brine (100 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, PE/EA = 10/1) to give S-500-6-1 \u2 (400mg, 67%) as a solid.
1 H NMR(400MHz,CDCl3)δ5.33-5.24(m,1H),4.06-4.00(m,1H),2.38-2.35(m,1H),2.08-1.82(m,6H),1.77-1.69(m,1H),1.62-1.20(m,13H),1.16-1.00(m,8H),0.99-0.92(m,1H),0.87-0.83(m,4H),0.74-0.64(m,3H)。
S-500-6-1 \/2 (350 mg) was purified by SFC (column:AD (250mm 30mm, 5um), conditions: 0.1% of NH 3 .H 2 O ETOH, gradient: from 35% to 35%, flow rate (ml/min): 60mL/min,25 ℃) to give 81 (peak 1, 130mg, 37%) and 62 (peak 2, 180mg, 52%) as solids.
81:
1 H NMR(400MHz,CDCl3)δ5.34-5.24(m,1H),4.09-4.00(m,1H),2.43-2.33(m,1H),2.14(d,J=4Hz,1H),2.07-1.80(m,5H),1.77-1.55(m,5H),1.53-1.30(m,7H),1.28-1.00(m,11H),1.00-0.91(m,1H),0.85(t,J=8Hz,3H),0.70(s,3H)。
LCMS Rt =1.220 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 25 H 38 F 3 O[M+H-H 2 O] + The calculated value 411 of (a), the measured value 411.
SFC peak 1: rt =4.561 min, 10 min chromatography, AD _3 \ u etoh _dea5 _40_25ml ("column: chiralpak AD-3 × 4.6mm i.d.,3um mobile phase: a: CO 2B: ethanol (0.05% dea) gradient: 5% to 40% B in 5 min and held at 40% for 2.5 min, then 5% B held for 2.5 min, flow rate: 2.5mL/min, column temperature: 35 ℃"), 100% de.
3. Pd (OH) 2 (0.2g,<1% water) was added to a solution of 81 (110mg, 0.256 mmol) in MeOH (2 mL) and THF (1 mL). The solution was hydrogenated under 50psi of hydrogen at 50 deg.C for 48 hours. The mixture was then filtered through a pad of celite and the filtrate was concentrated in vacuo. The residue was purified by flash column (PE/EtOAc =10/1 to 5/1) to give 56 (38mg, 35%) and 11 (42mg, 38%) as solids.
11:
1 H NMR(400MHz,CDCl3)δ4.09-3.99(m,1H),2.11(d,J=6.0Hz,1H),1.99-1.80(m,3H),1.70-1.55(m,6H),1.53-1.30(m,8H),1.27-0.96(m,11H),0.95-0.81(m,7H),0.70-0.61(m,4H)。
LCMS Rt =1.247 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 25 H 40 F 3 O[M+H-H 2 O] + Calculated value 413, found value 413.
Example 12: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -17- ((2S, 3R) -4- (4, 4-dimethylcyclohexyl) -3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (12)
Figure BDA0003762336660001141
Figure BDA0003762336660001151
1. Reacting NaBH 4 (2.80g, 82.5 mmol) was added five times every 5 minutes to a solution of S-500-6-29-u 2 (800mg, 1.65mmol) in MeOH (5 mL) and THF (5 mL). The mixture was stirred at 15 ℃ for 30 minutes. The mixture was washed with saturated NH 4 Cl (50 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases are washed with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-15% EtOAc in PE) to give S-500-6-30 (290mg, 36%) and 12 (120mg, 45%) as a solid.
12:
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.85-3.77(m,1H),2.40-2.32(m,1H),2.06-1.95(m,3H),1.77-1.58(m,7H),1.54-1.28(m,12H),1.27-1.06(m,11H),1.03(s,3H),1.00-0.95(m,2H),0.93-0.82(m,12H),0.69(s,3H)。
LCMS Rt =1.708 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 33 H 53 [M+H-2H 2 O] + Calculated 449, found 449.
Example 13: synthesis of (1S, 3R, 4S) -4- ((3S, 5S,8R,9S,10S,13S,14S, 17R) -3-hydroxy-10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-17-yl) -1-phenylpentane-1, 3-diol (13)
Figure BDA0003762336660001161
Figure BDA0003762336660001171
1. To a solution of N-4-1 \ (4) (27g, 85.8mmol) in THF (200 mL) were added CsF (25.9g, 171mmol) and TMSCF 3 (24.3g, 171mmol). The mixture was stirred at 10 ℃ for 1 hour. To the mixture was added water (10 mL) and TBAF.3H 2 O (30 g). The mixture was stirred at 30 ℃ for a further 2 hours. The mixture was concentrated in vacuo. The residue was dissolved in EtOAc (500 mL), washed with water (2X 500 mL), and washed with Na 2 SO 4 Dried, filtered, concentrated in vacuo and purified by flash column (DCM/EtOAc (1) in PE, 0-10%) to give N-4-1_5 (27g, 82%) and N-4-1_5a (3.5g, 11%) as a solid.
N-4-1_5:
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.70(s,1H),2.12-1.94(m,3H),1.89-1.78(m,2H),1.75(s,3H),1.72-1.60(m,5H),1.58-1.48(m,2H),1.45-1.09(m,10H),1.01-0.89(m,1H),0.85(s,3H),0.78-0.68(m,1H),0.56(s,3H)。
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.70(s,1H),2.09-1.99(m,1H),1.89-1.78(m,2H),1.75(s,3H),1.72-1.52(m,9H),1.45-1.06(m,10H),1.00-1.81(m,2H),0.79(s,3H),0.56(s,3H)。
2. At 40 ℃ in N 2 9-BBN dimer (29g, 119mmol) was added to a solution of N-4-1 \ (23g, 59.8mmol) in THF (250 mL) and stirred for 16 h. Ethanol (34.3mL, 598 mmol) and NaOH (119mL, 5M,598 mmol) were added to the reaction mixture. The mixture became clear. H is added dropwise at 25 DEG C 2 O 2 (59.8mL, 10M, 598mmol) and the internal temperature was raised to reflux (70 ℃ C.). After addition the mixture was cooled to 30 ℃. To the mixture was added Na 2 SO 3 (100mL, 20. The aq.). The organic layer was separated and poured into water (800 mL). A solid was formed. The mixture was filtered and the solid was washed with water, dried in vacuo and triturated with MeCN (250 mL) to give a solid. The solid was triturated from MeOH/water (250 mL/12.5 mL) at 60 deg.C and cooled to 15 deg.CFiltering at the temperature. The solid was vacuum dried to give N-4-1 \u6 (16.4 g, 68%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.69-3.60(m,1H),3.39-3.29(m,1H),2.09-2.01(m,1H),1.99-1.92(m,1H),1.87-1.75(m,2H),1.72-1.43(m,7H),1.42-1.07(m,11H),1.03(d,J=6.8Hz,3H),1.01-0.86(m,3H),0.85(s,3H),0.73-0.69(m,1H),0.67(s,3H)。
3. To a suspension of N-4-1 \ (u 6) (5g, 12.4mmol) in DCM (200 mL) was added water (223mg, 12.4mmol) and DMP (10.5g, 24.8mmol). The mixture was stirred at 15 ℃ for 15 minutes. The mixture is washed with NaHCO 3 /Na 2 S 2 O 3 (200 mL/200mL, sat.) twice, over Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-4-1_7 (4.5g, 90%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.60-9.51(m,1H),2.40-2.30(m,1H),2.12-1.78(m,5H),1.75-1.59(m,4H),1.57-1.15(m,11H),1.14-0.84(m,8H),0.78-0.63(m,5H)。
4. MeLi (7.75mL, 1.6M,12.4 mmol) was added to a solution of N-4-1 \u7 (1g, 2.49mmol) in THF (10 mL) at 0 ℃. The mixture was stirred at 15 ℃ for 1h. Adding NH to the mixture 4 Cl (10%, 20 mL). The mixture was extracted with EtOAc (2 × 30 mL). The combined organic layers were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a mixture (1 g) as a gum. The mixture N-3-2- \ u 1 (1 g) was purified by flash column (0-15% EtOAc in PE) to yield 91 (450 mg) and 22 (460 mg) and 130mg mixtures. 91 (450 mg) was recrystallized from MeCN (10 mL) to give 91 (50 mg) as a solid, and 22 (460 mg) was recrystallized twice from MeCN (10 mL) to give 22 (50 mg, as a solid).
91:
1 H NMR(400MHz,CDCl 3 )δ3.98-3.88(m,1H),2.11-2.02(m,1H),2.00(s,1H),1.98-1.88(m,2H),1.85-1.79(m,1H),1.73-1.58(m,4H),1.52-1.20(m,11H),1.19-1.11(m,4H),1.10-1.00(m,3H),0.97-0.89(m,4H),0.85(s,3H),0.75-0.68(m,1H),0.66(s,3H)。
LCMS Rt =1.155 min, chromatography at 2.0 min, 30-90_AB _E, 100% purity, MS ESI C 24 H 38 F 3 O[M+H-H 2 O] + Calculated value 399, found value 399.
HPLC Rt =5.23 min, chromatography at 10.0 min, 30-90 \uab _e, purity 98.88%, d.e.100%.
22:
1 H NMR(400MHz,CDCl 3 )δ3.97-3.82(m,1H),2.10-1.92(m,3H),1.85-1.78(m,1H),1.77-1.60(m,5H),1.59-1.06(m,13H),1.05-0.81(m,12H),0.74-0.62(m,4H)。
LCMS Rt =1.136 min, chromatography at 2.0 min, 30-90_AB _E, purity 100%, MS ESI C 24 H 38 F 3 O[M+H-H 2 O] + Calculated value 399, found value 399.
HPLC Rt =5.05 min, chromatography at 10.0 min, 30-90 \uab _e, purity 100%, d.e.100%.
5. To a solution of N-3-2 \ (u 1) (0.88g, 2.11mmol) in DCM (20 mL) was added water (2 drops) and DMP (1.78g, 4.22mmol). The mixture was stirred at 25 ℃ for 30 minutes. The mixture is washed with NaHCO 3 /Na 2 S 2 O 3 (30 mL/30mL, sat.) twice with Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-3-2_2 (0.85g, 97%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.53-2.42(m,1H),2.13-2.00(m,4H),1.97-1.78(m,2H),1.75-1.45(m,9H),1.43-1.13(m,9H),1.11(d,J=8.4Hz,3H),1.07-1.00(m,1H),0.98-0.88(m,1H),0.85(s,3H),0.78-0.68(m,1H),0.67(s,3H)。
6. To a solution of N-3-2 (0.85g, 2.05mmol) in DCM (5 mL) at 0 deg.C was added imidazole (279mg, 4.10mmol) and TMSCl (333mg, 3.07mmol). The mixture was stirred at 0 ℃ for 0.5h. The mixture was purified by addition of NaHCO 3 Quenched (20mL, sat) and extracted with PE (15 mL). Separating the organic layer with Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain N-3-2 \u2A (0)98g, 98%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.53-2.42(m,1H),2.13-2.03(m,4H),1.97-1.78(m,2H),1.75-1.31(m,11H),1.31-1.00(m,10H),1.00-0.88(m,1H),0.83(s,3H),0.75-0.61(m,4H),0.15(s,9H)。
7. BuLi (0.384mL, 2.5M, 0.615mmol) was added to i-Pr at-70 deg.C 2 A solution of NH (62.2 mg, 0.615mmol) in THF (0.5 mL) was stirred at 0 ℃ for 10 min. A solution of N-3-2_2A (0.2g, 0.41mmol) in THF (1 mL) was added at-70 ℃ and stirred for 30 min at-70 ℃. A solution of benzaldehyde (91.3mg, 0.861mmol) in THF (0.5 mL) was added at-70 ℃ and stirred for 15 min at-70 ℃. Reacting NH 4 Cl (1 mL, sat., aq.) was added to the mixture and extracted with EtOAc (10 mL). Separating the organic layer with Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-3-2_3c (250 mg, crude) as an oil.
1 H NMR(400MHz,CDCl 3 )δ7.46-7.30(m,5H),5.70-5.55(m,1H),3.60-3.25(m,1H),2.90-2.70(m,2H),2.55-2.41(m,1H),2.16-2.00(m,2H),1.96-1.75(m,3H),1.50-1.15(m,9H),1.13-1.05(m,4H),1.05-0.88(m,4H),0.87-0.80(m,5H),0.73-0.62(m,5H),0.15(s,9H)。
8. Subjecting LiAlH to 0 deg.C 4 (159mg, 4.20mmol) was added to a solution of N-3-2_3C (250mg, 0.421mmol) in THF (10 mL). The mixture was stirred at 0 ℃ for 5 minutes. Water (0.16 mL), naOH (0.16ml, 15 aq.) and water (0.48 mL) were added to the mixture in this order. The mixture was filtered and the solid was washed with THF (30 mL). The combined filtrates were concentrated in vacuo to give N-3-2_4C (250mg, 100%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ7.42-7.28(m,5H),5.70-5.30(m,1H),4.15-3.65(m,1H),2.18-1.55(m,9H),1.53-1.00(m,15H),1.00-0.75(m,9H),0.75-0.50(m,4H),0.15(s,9H)。
9. TBAF (219mg, 0.84mmol) was added to a solution of N-3-2_4C (250mg, 0.42mmol) in THF (2 mL). The mixture was stirred at 25 ℃ for 3h. The mixture was concentrated in vacuo. The residue was dissolved in EtOAc (10 mL) and washed with water (3 × 10 mL), purified by flash column (10-25% EtOAc in PE) to give N-3-2 \/5 c (150mg, 68%) as a solid.
10. The mixture N-3-2_5C (150 MG) was separated by SFC (instrument: MG-II; column: IC (250mm. 30mm, 10um); condition: 0.1% NH 3 H 2 O MeOH; and starting B:30 percent; and B, ending: 30 percent; flow rate (mL/min): 60, adding a solvent to the mixture; injecting: 300 To obtain impure 79 (35 mg, impure), a mixture of 31 and 25 (55 mg) and 13 (28 mg, impure).
Impure 79 (35 mg) was purified by flash column (10-30% EtOAc in PE) and the eluate was concentrated in vacuo. The residue was dissolved in MeCN/water (20ml, 4, 1) and concentrated in vacuo to give 79 (12 mg) as a solid.
25 and 13 (55 MG) were separated by SFC (instrument: MG-II; column: AD (250mm. About.30mm, 5 um); condition: 0.1% NH 3 H 2 O MeOH; and starting B:35 percent; and B, ending: 35 percent; flow rate (mL/min): 60; injecting: 70). Each eluate was separately concentrated in vacuo, dissolved in MeCN/water (20ml, 4.
Impure 31 (28 mg) was purified by SFC (instrument: SFC 17; column: AS (250mm. About. 30mm,5 um); conditions: 0.1% NH 3 H 2 O EtOH; and starting B:30 percent; and (5) finishing B:30 percent; flow rate (mL/min): 50; injecting: 60 To obtain a solid. The residue was dissolved in MeCN/water (20ml, 4) and concentrated in vacuo to give 31 (9 mg) as a solid.
SFC of four isomers peak 1: rt =1.501 minutes, peak 2: rt =1.730 minutes and peak 3: rt =1.943 min, IC-3 \ u MeOH (DEA) _40_2.5ML ("column: chiralPak IC-3X 4.6mm I.D.,3um; gradient: 40% methanol (0.05% DEA) in CO with 10 min chromatography 2 The preparation method comprises the following steps of (1) performing; flow rate: 2.5mL/min; column temperature: 40 deg.C ".
SFC of 25 and 13, peak 1: rt =4.411 minutes and peak 2: rt =4.920 min, 8min chromatography, AD _ MEOH (DEA) _5_40_2,8ML _8MIN ("column: chiralpak AD-3100X 4.6mm I.D.,3um; mobile phase: A: CO 2 B methanol (0.05% DEA); gradient: 5% to 40% by volume B within 4.5min and at 40% for 2.5 minutes,then 5% by weight B for 1min; flow rate: 2.8mL/min; column temperature: 40 deg.C ".
79:
1 H NMR(400MHz,CDCl 3 )δ7.43-7.28(m,5H),5.05-4.94(m,1H),4.04-3.91(m,1H),2.51(brs,1H),2.07-1.78(m,6H),1.70-1.61(m,4H),1.51-1.41(m,3H),1.39-1.12(m,11H),1.05-0.98(m,2H),0.91-0.81(m,7H),0.71-0.60(m,4H)。
LCMS Rt =1.298 min, chromatography over 2min, 10-80AB _2MIN _E, purity 96.7%, MS ESI C 31 H 45 F 3 O 3 Na[M+Na] + Calculated value 545, found value 545.
SFC Rt =1.483 min, IC-3 \ u MeOH (DEA) _40 _u2.5ML in 10 min chromatography, 100% de.
25:
1 H NMR(400MHz,CDCl 3 )δ7.42-7.28(m,5H),4.97-4.81(m,1H),4.12-3.92(m,1H),3.23(brs,1H),2.69(brs,1H),2.10-1.88(m,3H),1.82-1.62(m,7H),1.48-1.18(m,10H),1.10-0.88(m,8H),0.87-0.78(m,4H),0.70-0.58(m,4H)。
LCMS Rt =1.319 min, chromatography over 2min, 10-80AB _2MIN _E, purity 97.0%, MS ESI C 31 H 45 F 3 O 3 Na[M+Na] + Calculated value 545, found value 545.
SFC Rt =1.718 min, IC-3 \ u MeOH (DEA) _40_2.5ML,98.26% by 5min chromatography.
SFC Rt =4.367 min, 8min chromatography, AD _ MEOH (DEA) _5_40_2,8ML _8MIN,100% de.
31:
1 H NMR(400MHz,CDCl 3 )δ7.45-7.28(m,5H),5.02-4.81(m,1H),4.18-3.98(m,1H),3.35(brs,1H),2.47(brs,1H),2.15-1.72(m,8H),1.53-1.31(m,8H),1.30-1.03(m,8H),0.99-0.89(m,4H),0.89-0.78(m,4H),0.75-0.60(m,4H)。
LCMS Rt =1.327 min, chromatography over 2min, 10-80AB _2MIN _E, 100% purity, MS ESI C 31 H 45 F 3 O 3 Na[M+Na] + Calculated value 545, found value 545.
SFC Rt =1.929 min, IC-3 \ u MeOH (DEA) _40 _u2.5ML, 98.4% de by 10 min chromatography.
13:
1 H NMR(400MHz,CDCl 3 )δ7.40-7.28(m,5H),5.12-5.07(m,1H),3.95-3.88(m,1H),2.76(brs,1H),2.08-1.78(m,6H),1.75-1.60(m,5H),1.51-1.38(m,4H),1.36-1.09(m,9H),1.00-0.89(m,6H),0.83(s,3H),0.71-0.64(m,1H),0.63(s,3H)。
LCMS Rt =1.309 min, chromatography over 2min, 10-80AB _2MIN _E, purity 100%, MS ESI C 31 H 45 F 3 O 3 Na[M+Na] + Calculated value 545, found value 545.
SFC Rt =1.683 min, IC-3 \ u MeOH (DEA) _40 _u2.5ML, 98.94% de by chromatography over 5 min.
SFC Rt =4.785 min, 8min chromatography, AD _ MEOH (DEA) _5_40_2,8ML _8MIN, 94.03%.
Example 14: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R, E) -3-hydroxy-5-phenylpent-4-en-2-yl) -10, 13-dimethyl-3- (trifluoromethyl) hexadec-hydro-1H-cyclopenta [ a ] phenanthren-3-ol (14)
Figure BDA0003762336660001231
Figure BDA0003762336660001241
1. BuLi (0.384mL, 2.5M, 0.615mmol) was added to i-Pr at-70 deg.C 2 A solution of NH (62.2mg, 0.615mmol) in THF (0.5 mL) was stirred at 0 ℃ for 10 min. A solution of N-3-2_2A (0.2g, 0.41mmol) in THF (1 mL) was added at-70 ℃ and stirred at-70 ℃ for 1h. A solution of benzaldehyde (91.3mg, 0.861mmol) in THF (0.5 mL) was added at-70 ℃ and stirred at 20 ℃ for 4h. Reacting NH 4 Cl (1mL, sat., aq.) was added to the mixture and washed with EtOAc (10 mL) extraction. Separating the organic layer with Na 2 SO 4 Dried, filtered and concentrated in vacuo, purified by flash column (0-10% EtOAc in PE) to give N-3-2_3D (150mg, 64%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ7.64-7.55(m,3H),7.43-7.39(m,3H),6.79(d,J=16.0Hz,1H),2.86-2.73(m,1H),2.15-2.08(m,1H),2.00-1.90(m,1H),1.88-1.80(m,1H),1.72-1.59(m,5H),1.53-1.22(m,9H),1.21-1.03(m,7H),0.99-0.88(m,1H),0.84(s,3H),0.75-0.61(m,4H),0.15(s,9H)。
2. TBAF (135mg, 0.52mmol) was added to a solution of N-3-2_3D (150mg, 0.26mmol) in THF (1 mL). The mixture was stirred at 20 ℃ for 20h. To the mixture was added EtOAc (5 mL). The mixture was washed with water (2 × 5 mL), brine (5 mL) and concentrated in vacuo to afford N-003-005_1 (140 mg, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ7.64-7.53(m,3H),7.43-7.35(m,3H),6.79(d,J=16.0Hz,1H),2.88-2.73(m,1H),2.13-1.90(m,3H),1.88-1.78(m,1H),1.77-1.90(m,5H),1.60-1.22(m,8H),1.21-0.88(m,9H),0.86(s,3H),0.75-0.61(m,4H)。
3. NaBH is reacted at 20 ℃ 4 (419mg, 11.1mmol) was added portionwise to a solution of N-003-005_1 (140mg, 0.278mmol) in THF (2 mL) and MeOH (1 mL). The mixture was stirred at 20 ℃ for 10 minutes. The reaction was performed using water (20 mL) and NH 4 Cl (20mL, sat.). The mixture was extracted with EtOAc (50 mL). The organic layer was concentrated in vacuo and purified by prep-TLC (PE/EtOAc = 4/1) to give N-003-005 (50 mg, impure) and N-003-006 (50 mg), both as solids.
14 (50 mg) was dissolved in MeCN (20 mL) and concentrated in vacuo to give 14 (29 mg) as a solid.
14:
1 H NMR(400MHz,CDCl 3 )δ7.44-7.38(m,2H),7.37-7.29(m,2H),7.25-7.18(m,1H),6.59(d,J=16.0Hz,1H),6.24(dd,J=7.2,16.0Hz,1H),4.40-4.30(m,1H),2.08-1.92(m,3H),1.89-1.77(m,3H),1.68-1.60(m,3H),1.50-1.08(m,13H),1.03-0.82(m,9H),0.72-0.62(m,4H)。
LCMS Rt =1.236 min, chromatography over 2min, 30-90AB _2MIN _E, 99.0% purity, MS ESI C 31 H 42 F 3 O[M+H-H 2 O] + Calculated value 487 of (a), measured value 487.
HPLC Rt =5.89 min, chromatography for 8 min, 30-90_AB _ _1.2ml,98.1%
Example 15: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-6, 6-dimethylhept-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (15)
Figure BDA0003762336660001251
Figure BDA0003762336660001261
1. Pd (OH) 2 the/C (100 mg) was added to a solution of 44 (100 mg) in MeOH/THF (2 mL/2 mL). The mixture was heated at 50 ℃ in H 2 Stirring was carried out for 20h at 50 psi. The mixture was filtered. The filtrate was concentrated to give 100mg of solid. NMR showed 9%54 retention. The impure sample was hydrogenated 3 more times under the same conditions. The mixture was filtered. The filtrate was concentrated and separated by flash column (0-15% etoac in PE) to give 5 (7 mg) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.66-3.48(m,1H),2.00-1.55(m,9H),1.50-1.22(m,15H),1.19-1.03(m,8H),0.96(s,3H),0.91-0.81(m,15H),0.67(s,3H)。
LCMS Rt =1.492 minutes, chromatography at 2.0 minutes, 30-90_AB _E, 100% purity, MS ESI C 30 H 51 [M+H-2H 2 O] + The calculated value 411 of (a), the measured value 411.
Example 16: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-10, 13-dimethyl-17- ((2S, 3R) -4, 4-trifluoro-3-hydroxybutan-2-yl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (16)
Figure BDA0003762336660001262
1. Pd (OH) 2 (0.2g,<1% water) was added to a solution of 62 (160mg, 0.373mmol) in MeOH (2 mL) and THF (1 mL). The solution was hydrogenated under 50psi hydrogen at 50 ℃ for 16 hours. The mixture was then filtered through a pad of celite and the filtrate was concentrated in vacuo. The residue was purified by flash column (PE/EtOAc =10/1 to 5/1) to give 44 (27mg, 17%) and 16 (117mg, 73%) as solids.
16:
1 H NMR(400MHz,CDCl3)δ4.04-3.96(m,1H),1.98-1.83(m,4H),1.69-1.59(m,3H),1.56-1.20(m,13H),1.17-0.95(m,8H),0.91-0.83(m,8H),0.70-0.62(m,4H)。
LCMS Rt =1.240 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 25 H 40 F 3 Calculated value of O [ M + H-H ] 2 O] + 413, found value 413.
Example 17: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-6, 6-dimethylhept-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (17)
Figure BDA0003762336660001271
1. Pd (OH) 2 the/C (100 mg) was added to a solution of 5 (250 mg) in MeOH/THF (2 mL/2 mL). The mixture was heated at 50 ℃ in H 2 Stirring was continued for 20 hours at 50 psi. The mixture was filtered. The filtrate was concentrated to give 250mg of a solid. NMR showed 70%5 remaining. The impure sample was hydrogenated 3 more times under the same conditions. The mixture was filtered. The filtrate was concentrated and separated by flash column (0-15% etoac in PE) to give 17 (3 mg) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.63-3.50(m,1H),1.98-1.55(m,8H),1.49-1.37(m,8H),1.35-1.21(m,8H),1.19-1.01(m,8H),0.97(s,3H),0.91-0.82(m,15H),0.66(s,3H)。
LCMS Rt =1.529 min, chromatography at 2.0 min, 30-90 u AB _E, purity 95.6%, MS ESI C 30 H 51 [M+H-2H 2 O] + The calculated value 411 of (a), the measured value 411.
Example 18: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-4- (tetrahydro-2H-pyran-4-yl) but-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (18)
Figure BDA0003762336660001281
1. To Me at 20 ℃ under nitrogen 3 To a suspension of SI (8.44g, 41.4 mmol) in dry THF (50 mL) was added t-BuOK (4.64g, 41.4 mmol). The mixture was stirred at 20 ℃ for 1 hour, and N-005_5 (5g, 13.8mmol) was added. The resulting mixture was warmed to 45 ℃ and stirred for 4 hours. The reaction mixture was cooled to room temperature, quenched with water (50 mL) and extracted with EtOAc (2 × 50 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography on silica gel (0-10% etoac in PE) to give N-005_001 (2.7g, 52%) as a solid.
LCMS Rt =1.324 min, chromatography at 2.0 min, 30-90AB _E, 92% purity, MS ESI C 25 H 41 O[M+H-H 2 O] + Calculated value 357, found value 357.
2. In N 2 A solution of 4-chlorotetrahydro-2H-pyran (1g, 8.29mmol) in dry THF (8 mL) was added dropwise to Mg (401mg, 16.5 mmol) and I at 60 deg.C 2 (105mg, 0.414mmol) in 2mL dry THF). The mixture was stirred at 60 ℃ for 10 minutes. The temperature was increased to 66 ℃. The reaction mixture was stirred for an additional 30 minutes, cooled to room temperature and used directly as a solution of (tetrahydro-2H-pyran-4-yl) magnesium chloride (0.83M in THF).
3. A solution of (tetrahydro-2H-pyran-4-yl) -magnesium chloride (0.83M in THF, 6.38mL, 5.30mmol) was added dropwise to N-005 _0at 20 deg.C under nitrogen01 (400mg, 1.06mmol) and CuI (20.1mg, 0.106mmol) in dry THF (10 mL). The mixture was stirred at 20 ℃ for 18 hours. The reaction mixture was washed with water (10 mL) and saturated NH 4 Cl (10 mL) was quenched and extracted with EtOAc (2X 15 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give N-6-9/10 (760 mg, crude) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.01-3.92(m,3H),3.83-3.62(m,1H),3.42-3.32(m,3H),1.97-1.87(m,1H),1.68-1.58(m,7H),1.57-1.45(m,6H),1.43-1.29(m,8H),1.24-0.95(m,10H),0.91-0.79(m,9H),0.73-0.56(m,4H)。
LCMS Rt =1.332 min, chromatography 2.0 min, 10-80AB, 93% purity, MS ESI C 30 H 52 NaO 3 [M+Na] + Calculated value 483, found value 483.
4. BzCl (691mg, 4.92mmol) and DMAP (20mg, 0.164mmol) were added to a solution of N-6-9/10 (760mg, 1.64mmol) in pyridine (10 mL). The mixture was stirred at 20 ℃ for 2 hours. The reaction mixture was quenched with water (15 mL) and extracted with EtOAc (2 × 20 mL). The combined organic layers were diluted with 10% aqueous HCl (2X 20 mL), saturated NaHCO 3 Washed (40 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography on silica gel (0-10% EtOAc in PE) to give N-6-9 \u1 (400mg, 43%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ8.08-8.01(m,1H),8.08-8.01(m,1H),7.61-7.42(m,3H),5.42-5.29(m,1H),3.99-3.85(m,2H),3.41-3.24(m,2H),2.06-1.65(m,5H),1.65-1.57(m,5H),1.54-1.42(m,6H),1.42-1.14(m,11H),1.14-0.90(m,8H),0.89-0.77(m,7H),0.69-0.51(m,4H)。
N-6-9_1 (400mg, 0.708mmol) was isolated and purified by SFC (column: C2 250mm 30mm,10um, gradient: 45-45% 3 /H 2 O, B = MeOH), flow rate: 60 mL/min) to yield solid N-6-9 \u2 (peak 1, rt =3.926 min, 80mg, 20%) and solid N-6-10 \u1 (peak 2, rt =4.893 min, 180mg, 45%).
N-6-9_2:
1 H NMR(400MHz,CDCl 3 )δ8.06-8.00(m,2H),7.60-7.53(m,1H),7.49-7.42(m,2H),5.35-5.28(m,1H),3.98-3.91(m,2H),3.41-3.31(m,2H),1.88-1.67(m,5H),1.66-1.57(m,4H),1.54-1.36(m,10H),1.35-1.16(m,8H),1.08-0.88(m,8H),0.88-0.82(m,4H),0.80(s,3H),0.64(s,3H),0.61-0.54(m,1H)。
LCMS Rt =1.540 min, chromatography at 2.0 min, 30-90AB, 96% purity, MS ESI C 30 H 49 O[M-BzOH-H 2 O+H] + The calculated value 425 and the measured value 425 of (c).
SFC Rt =3.789 min, chromatography over 8 min, column: lux Cellulose-2X 4.6mm I.D.,3 μm; mobile phase: 40% methanol (0.05% 2 Performing the following steps; flow rate: 2.5mL/min; column temperature: 40 deg.C, 97% de.
N-6-10_1:
1 H NMR(400MHz,CDCl 3 )δ8.06-8.01(m,2H),7.61-7.53(m,1H),7.49-7.42(m,2H),5.41-5.33(m,1H),3.98-3.86(m,2H),3.40-3.27(m,2H),2.05-1.91(m,2H),1.84-1.72(m,2H),1.66-1.59(m,3H),1.55-1.38(m,9H),1.37-1.16(m,11H),1.13-1.00(m,6H),1.00-0.90(m,2H),0.89-0.79(m,7H),0.67(s,3H),0.63-0.54(m,1H)。
LCMS Rt =1.507 min, chromatography at 2.0 min, 30-90AB, 97% purity, MS ESI C 30 H 49 O[M-BzOH-H 2 O+H] + The calculated value 425 and the measured value 425 of (c).
SFC Rt =4.699 min, at 8 min chromatography, column: lux Cellulose-2X 4.6mm I.D.,3 μm; mobile phase: 40% methanol (0.05% 2 The preparation method comprises the following steps of (1) performing; flow rate: 2.5mL/min; column temperature: 40 deg.C, 97% de.
5. Water (1 mL) and KOH (78.5mg, 1.40mmol) were added to a solution of N-6-9 \u2 (80mg, 0.141mmol) in THF (2 mL) and methanol (1 mL). The mixture was stirred at 50 ℃ for 18 hours. The reaction mixture was cooled, diluted with water (5 mL), acidified with 10% hcl (0.2 mL) and extracted with EtOAc (3 × 5 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography on silica gel (10-30% etoac in PE) to give 18 (13mg, 20%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.05-3.89(m,3H),3.45-3.34(m,2H),1.94-1.87(m,1H),1.86-1.76(m,1H),1.72-1.59(m,6H),1.54-1.45(m,4H),1.44-1.28(m,9H),1.28-1.15(m,7H),1.13-0.92(m,5H),0.91-0.85(m,4H),0.84-0.79(m,6H),0.70-0.62(m,4H)。
LCMS Rt =1.213 min, chromatography at 2.0 min, 30-90AB, purity 100%.
MS MS ESI C 30 H 49 O[M-2H 2 O+H] + The calculated value 425 and the measured value 425 of (c).
Example 19: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((1S, 2S) -1-hydroxy-1-phenylprop-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (19)
Figure BDA0003762336660001311
1. A solution of N-8-7_1 (300mg, 0.832mmol) in THF (5 mL) was added to a solution of PhMgBr (1.38mL, 3M in ether, 4.15 mmol) in THF (10 mL) at 0 deg.C, and the reaction mixture was stirred at 0 deg.C for 3 hours. Then, the reaction mixture was stirred at 25 ℃ for 5 hours. The reaction mixture was quenched by water (10 mL) at 0 ℃. The solution was filtered and the filter cake was washed with EtOAc (10 mL). The aqueous phase was extracted with EtOAc (3X 15 mL). The combined organic phases were washed with saturated brine (2X 10 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE/EtOAc =20/1 to 1/1) to give the crude product (200 mg) as a solid. The crude product was purified by SFC (column: AD (250mm 30mm, 5um)), gradient: 25-25% B (a =0.1% nh3/H2O, B = EtOH), flow rate: 60 mL/min) to give 55 (Peak 2, 55mg, 15%) and 19 (Peak 1, 21mg, 6%) as solids.
19:
1 H NMR(400MHz,CDCl 3 )δ7.40-7.20(m,5H),4.85-4.80(m,1H),2.10-1.60(m,5H),1.55-1.05(m,17H),0.95-0.75(m,14H),0.71(s,3H),0.60-0.50(m,1H)。
LCMS Rt =1.208 min, chromatography at 2.0 min, 30-90ab \u2 min. Purity 100%, MS ESI C 30 H 43 Calculated value of [ M-2H ] 2 O+H] + 403, measured value 403.
SFC Rt =1.047 min, OJ _ 3. Mu. EtOH _DEA _. Mu. 5. Mu. 40. Mu. 25ML,100% by 3 min chromatography.
Example 20: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -4- (4, 4-dimethylcyclohexyl) -3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (20)
Figure BDA0003762336660001321
Figure BDA0003762336660001331
1. Pd (OH) 2 (150 mg, anhydrous) was added to a solution of 12 (100mg, 0.206mmol) in MeOH (20 mL). The mixture was heated at 50 ℃ in H 2 Next (50 Psi) was stirred for 48 hours. The mixture was filtered, concentrated and purified by combi-flash (0-15% EtOAc in PE) to give 87 (12mg, 12%) and 20 (11mg, 11%) as solids.
20:
1 H NMR(400MHz,CDCl 3 )δ3.83-3.75(m,1H),1.99-1.92(m,1H),1.71-1.57(m,8H),1.52-1.43(m,3H),1.41-1.29(m,8H),1.27-1.13(m,11H),1.12-1.04(m,4H),1.03-0.94(m,3H),0.91-0.86(m,12H),0.82(s,3H),0.68-0.59(m,4H)。
LCMS Rt =1.748 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 33 H 55 [M+H-H 2 O] + Calculated value 451 of (d), found value 451.
Example 21: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxypentan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (21)
Figure BDA0003762336660001332
Figure BDA0003762336660001341
1. At 25 ℃ in N 2 EtMgBr (0.553mL, 3M in ether, 1.66 mmol) was then added dropwise to a solution of N-8_1 (250mg, 0.8320mmol) in THF (3 mL). The mixture was stirred at 25 ℃ for 1 hour with saturated NH 4 Cl (10 mL) quenched and extracted with EtOAc (3 × 15 mL). The organic layer was washed with brine (20 mL) and Na 2 SO 4 Dried, filtered, and concentrated in vacuo to give the crude product N-8-24_1, which was purified by flash column (0-15% EtOAc in PE) to give 21 (130 mg, impure) as a solid. Impure N-8-24 (130mg, 0.3327mmol) was recrystallized from MeCN (3 mL) at 85 ℃ to give pure 21 (111mg, 86%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.62-3.50(m,1H),2.02-1.81(m,2H),1.72-1.59(m,3H),1.56-1.46(m,4H),1.45-1.17(m,12H),1.16-1.00(m,5H),0.99-0.85(m,11H),0.84-0.78(m,4H),0.66(s,4H)。
HPLC Rt =5.73 min, chromatography over 10 min, 30-90_ab _1.2ml _, purity 100%.
MS MS ESI C 26 H 43 [M+H-2H 2 O] + The calculated value 355, found value 355.
Example 22: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -3-hydroxybut-2-yl) -10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (22)
Figure BDA0003762336660001351
1. At 0 deg.CMeLi (7.75mL, 1.6M, 12.4mmol) was added to a solution of N-4-1 \7 (1g, 2.49mmol) in THF (10 mL). The mixture was stirred at 15 ℃ for 1 hour. Adding NH to the mixture 4 Cl (10%, 20 mL). The mixture was extracted with EtOAc (2 × 30 mL). The combined organic layers were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a mixture (1 g) as a gum. The mixture (1 g) was purified by flash column (0-15% EtOAc in PE) to yield 91 (450 mg) and 22 (460 mg) and 130mg mixtures. 91 (450 mg) was recrystallized from MeCN (10 mL) to give 91 (50 mg) as a solid. 22 (460 mg) was recrystallized twice from MeCN (10 mL) to give 22 (50 mg) as a solid.
22:
1 H NMR(400MHz,CDCl 3 )δ3.97-3.82(m,1H),2.10-1.92(m,3H),1.85-1.78(m,1H),1.77-1.60(m,5H),1.59-1.06(m,13H),1.05-0.81(m,12H),0.74-0.62(m,4H)。
LCMS Rt =1.136 min, chromatography at 2.0 min, 30-90_AB _E, 100% purity, MS ESI C 24 H 38 F 3 O[M+H-H 2 O] + Calculated value 399, actually measured value 399.
HPLC Rt =5.05 min, chromatography at 10.0 min, 30-90_ab \ue, purity 100%, d.e.100%.
Example 23: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3-ethyl-17- ((2S, 3S) -3-hydroxyhex-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (23)
Figure BDA0003762336660001361
1. Propylmagnesium bromide (3.34mL, 6.69mmol,2M in THF) was slowly added to a solution of S-500-6-1 \1 (800mg, 2.23mmol) in THF (30 mL) at 0 deg.C. After the addition, the mixture was stirred at 15 ℃ for 1 hour. Mixing with sat 4 Cl (40 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases were washed with brine (2X 30 mL) and Na 2 SO 4 Dried, filtered and concentrated and purified by combi-flash (0-15% EtOAc in PE) to give 72 (500 m)g, 56%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.72-3.64(m,1H),2.41-2.31(m,1H),2.07-1.85(m,4H),1.77-1.69(m,1H),1.62-1.54(m,3H),1.52-1.38(m,9H),1.37-1.16(m,6H),1.15-1.01(m,7H),0.99-0.88(m,7H),0.87-0.82(m,3H),0.68(s,3H)。
LCMS Rt =4.979 min, chromatography at 7.0 min, 30-90AB _E, 98.8% purity, MS ESI C 27 H 43 [M+H-2H 2 O] + The calculated value 367 and the actual value 367 of (a).
2. Pd (OH) 2 (300 mg, anhydrous) was added to a solution of 72 (150mg, 0.372mmol) in MeOH (20 mL). The mixture was heated at 50 ℃ in H 2 Next (50 Psi) was stirred for 48 hours. The mixture was filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 23 (9mg, 6%) and 38 (43mg, 29%) as solids.
23:
1 H NMR(400MHz,CDCl 3 )δ3.71-3.62(m,1H),2.01-1.83(m,3H),1.82-1.72(m,1H),1.69-1.57(m,3H),1.51-1.37(m,9H),1.36-1.22(m,9H),1.20-1.00(m,8H),0.97(s,3H),0.94-0.87(m,8H),0.66(s,3H)。
LCMS Rt =1.440 min, chromatography at 2.0 min, 30-90AB _E, 98.8% purity, MS ESI C 27 H 45 [M+H-2H 2 O] + Found 369, found 369.
Example 24: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (24)
Figure BDA0003762336660001371
1. A solution of 1-bromo-3-methylbutane (11.7 g, 78mmol) in THF (8 mL) was added dropwise to Mg (4.35g, 179mmol) and I at 60 deg.C 2 (20 mg) in THF (2 mL). The mixture was stirred at 60 ℃ for 1 hour. Will be mixed withThe compound was diluted with THF (10 mL) and used as received. At N 2 Freshly prepared isopentylmagnesium bromide (19.5mL, 3.9M in THF, 76 mmol) was added to a solution of S-200-INT-5E (1.0 g, 2.78mmol) in THF (5 mL) at 0 deg.C. The mixture was stirred at 0 ℃ for 1 hour. NH (NH) 4 Cl (20 mL, saturated aqueous solution) was added to the mixture. The mixture was extracted with EtOAc (2 × 30 mL). The combined organic phases were washed with brine (100 mL) and Na 2 SO 4 Dried, concentrated in vacuo, purified by silica gel (PE/EtOAc =20/1 to 10/1), and purified from CH 3 CN (10 mL) recrystallized as 77 (255mg, 21%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.32-5.26(m,1H),3.66-3.59(m,1H),2.42-2.32(m,1H),2.07-1.85(m,4H),1.77-1.58(m,4H),1.55-1.38(m,10H),1.38-1.19(m,5H),1.19-1.00(m,8H),1.00-0.81(m,13H),0.69(s,3H)。
LCMS Rt =1.306 min, chromatography at 2.0 min, 30-90 AB, 100% purity, MS ESI C 29 H 49 O[M+H-H 2 O] + Calculated value 413, found value 413.
2. At 25 ℃ in N 2 Benzoic acid (508mg, 4.16mmol) and triphenylphosphine (1.63g, 6.24mmol) were added to a solution of 77 (900mg, 2.08mmol) in THF (30 mL). The mixture was stirred for 20mins at 25 ℃. At 0 ℃ in N 2 Next, DIAD (1.26g, 6.24mmol) was added. The mixture was stirred at 0 ℃ for 20mins, warmed to 25 ℃ and stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (60 mL) and extracted with MTBE (3 × 30 mL). The combined organic phases were washed with brine (60 mL) and Na 2 SO 4 Dried, filtered, concentrated in vacuo to give the crude product, which was purified by flash column (0-10% etoac in PE) to give the impure product S-500-2-15 \u1 (900 mg) as an oil, which was used directly in the next step.
3. NaOH solution (974 mg in 6mL H 2 O, 16.8 mmol) was added to a solution of S-500-2-15_1 (900mg, 1.68mmol) in THF (10 mL) and MeOH (5 mL). The mixture was heated at 50 ℃ for 16 hours. Reaction mixture with saturated NH 4 Cl (60 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases are washed with brine (60)mL) and washed with Na 2 SO 4 Dried, filtered and concentrated, and purified by combi-flash (0-15% etoac in PE) to give 210mg solid, which was purified by SFC (column: AD (250mm × 30mm,5 um), gradient: 35-35% b (a =0.1% nh% 3 /H 2 O, B = MeOH), flow rate: 80 mL/min) to give 52 (150mg, 68%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.30-5.26(m,1H),3.64-3.58(m,1H),2.40-2.30(m,1H),2.02-1.92(m,3H),1.80-1.58(m,7H),1.56-1.31(m,9H),1.30-1.05(m,8H),1.03(s,3H),1.02-0.96(m,2H),0.95-0.86(m,9H),0.85-0.80(m,3H),0.69(s,3H)。
LCMS t R =1.335 min, chromatography by 2 min, 30-90AB _ELSD, purity 100.0%, MS ESI C 29 H 47 [M+H-2H 2 O] + 395, found 395.
4. Pd (OH) 2 (200 mg) was added to a solution of 52 (50mg, 0.116mmol) in MeOH (10 mL). The mixture was heated at 50 ℃ in H 2 Stirring was performed under (50 Psi). The mixture was filtered, concentrated and purified by combi-flash (0-10% etoac in PE) to give solid 24 (15mg, 30%) and solid 96 (1.2mg, 3%).
24:
1 H NMR(400MHz,CDCl 3 )δ3.66-3.52(m,1H),2.02-1.91(m,1H),1.74-1.57(m,7H),1.52-1.44(m,2H),1.43-1.29(m,7H),1.28-1.04(m,11H),1.03-0.94(m,3H),0.94-0.85(m,13H),0.82(s,3H),0.71-0.60(m,4H)。
LCMS t R =1.342 min, chromatography over 2 min, 30-90ab _elsd, purity 100.0%, MS ESI C 29 H 49 [M+H-2H 2 O] + Calculated value 397 of (a), found value 397.
Example 25: synthesis of (1S, 3S, 4S) -4- ((3S, 5S,8R,9S,10S,13S,14S, 17R) -3-hydroxy-10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-17-yl) -1-phenylpentane-1, 3-diol (25)
Figure BDA0003762336660001391
25 can be seen in example 13.
25:
1 H NMR(400MHz,CDCl 3 )δ7.42-7.28(m,5H),4.97-4.81(m,1H),4.12-3.92(m,1H),3.23(brs,1H),2.69(brs,1H),2.10-1.88(m,3H),1.82-1.62(m,7H),1.48-1.18(m,10H),1.10-0.88(m,8H),0.87-0.78(m,4H),0.70-0.58(m,4H)。
LCMS Rt =1.319 min, chromatography over 2min, 10-80AB _2MIN _E, purity 97.0%, MS ESI C 31 H 45 F 3 O 3 Na[M+Na] + Calculated value 545, found value 545.
SFC Rt =1.718 min, IC-3 \ u MeOH (DEA) _40_2.5ML,98.26% by 5 min chromatography.
SFC Rt =4.367 min, 8min chromatography, AD _ MEOH (DEA) _5 \u40 _2,8ML _, 8MIN, 100%.
Example 26: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((S) -3-hydroxy-3-methylbutan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (26)
Figure BDA0003762336660001401
1. At 0 ℃ in N 2 MeMgBr (0.83mL, 2.49mmol,3M in ether) was added dropwise to a solution of N-8-7 \u1 (300mg, 0.832 mmol) in THF (20 mL). After stirring for 30 minutes at 20 ℃ the reaction mixture was saturated with NH 4 Cl (50 mL) quenched and extracted with EtOAc (2 × 10 mL). The combined layers were washed with brine (10 mL) and Na 2 SO 4 Dried, filtered and concentrated to give a residue which was purified by flash column (0-10% etoac in PE) to give N-8-22_1 (100mg, 31%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.99-3.88(m,1H),1.98-1.84(m,2H),1.69-1.57(m,6H),1.52-1.45(m,2H),1.44-1.28(m,3H),1.26-1.17(m,5H),1.16-1.11(m,5H),1.10-0.95(m,5H),0.93-0.86(m,7H),0.84-0.80(m,4H),0.69-0.62(m,4H)。
2. DMP (224mg, 0.53mmol) was added to a solution of N-8-22/1 (100mg, 0.265mmol) in DCM (10 mL). After stirring for 10 min at 20 ℃ the reaction mixture was saturated with NaHCO 3 The solution (30 mL) was quenched until the pH of the aqueous layer was about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (20 mL). The combined organic phases were saturated with Na 2 S 2 O 3 (3x40mL)、sat.NaHCO 3 (40 mL), brine (40 mL), washed with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-20% etoac in DCM) to give N-8-22_2 (80mg, 80%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.54-2.42(m,1H),2.09(s,3H),1.94-1.87(m,1H),1.71-1.59(m,4H),1.54-1.45(m,3H),1.44-1.30(m,4H),1.29-1.16(m,6H),1.15-1.07(m,5H),1.06-0.92(m,4H),0.91-0.79(m,7H),0.74-0.61(m,4H)。
3. In N 2 MeMgBr (0.353mL, 1.06mmol,3M in diethyl ether) was added to a solution of N-8-22 \u2 (80mg, 0.213mmol) in THF (5 mL). After stirring for 30 minutes at 20 ℃ the reaction mixture is saturated with NH 4 Aqueous Cl (30 mL) was quenched and extracted with EtOAc (20mL. Times.3). The combined organic layers were washed with brine (50 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product, which was purified by silica gel column (0-10% etoac in PE) to give 26 (7 mg, 8%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.08-2.01(m,1H),1.97-1.86(m,1H),1.69-1.57(m,6H),1.53-1.45(m,3H),1.40-1.27(m,5H),1.26-1.17(m,8H),1.14(s,3H),1.13-1.01(m,3H),0.99-0.92(m,5H),0.91-0.85(m,4H),0.82(s,3H),0.70(s,3H),0.67-0.60(m,1H)。
LCMS Rt =1.240 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 26 H 43 Calculated value of [ M + H-2H ] 2 O] + 355, found 355.
Example 27: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-ethyl-17- ((2S, 3R) -3-hydroxy-4-methylpentan-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (27)
Figure BDA0003762336660001421
1. At 0 ℃ in N 2 To a solution of N-014-012 u 4 (300mg, 0.8366mmol) in THF (20 mL) was added dropwise isopropyl magnesium chloride solution (1.25mL, 2.50mmol, 2M) over 30mins, during which time the temperature was kept below 0 ℃. The reaction mixture was stirred at 20 ℃ for an additional 2 hours to give a suspension. Adding saturated NH to the reaction mixture 4 Aqueous Cl (15 mL) and stirred for 20 min, then the mixture was extracted with EtOAc (3 × 10 mL). The combined organic phases were washed with brine (2X 10 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated to give N-014-001_1 (360 mg, crude) as a solid, 1 h NMR showed the desired product and was used directly in the next step.
2.X1 (150mg, 0.37mmol) was purified by SFC (column: chiralpak AS-H250 × 305u; conditions: 0.1% NH 3 H 2 O EtOH; and starting B:20 percent; and (5) finishing B:20 percent; flow rate (ml/min): 65 To give 37 (peak 2, 46mg, 31%) and 27 (peak 1, 27mg, 18%) as solids.
37:
27:
1 H NMR(400MHz,CDCl 3 )δ5.32-5.26(m,1H),3.42-3.34(m,1H),2.43-2.34(m,1H),2.06-1.91(m,3H),1.90-1.75(m,2H),1.74-1.66(m,2H),1.63-1.58(m,3H),1.54-1.26(m,11H),1.22-1.04(m,3H),1.03-0.99(m,3H),0.97-0.93(m,7H),0.92-0.87(m,3H),0.86-0.77(m,3H),0.70(s,3H)。
LCMS Rt =1.228 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, MS ESI C 27 H 45 O[M+H-H 2 O] + The calculated value 385 of (1), the measured value 385.
SFC Rt =2.440 min, 10 min chromatography, OJ _ 3u EtOH _DEA _5 _u40 _25ML ("column: chiralcel OJ-3X 4.6mm I.D.; 3um mobile phase: A: CO 2B: ethanol (0.05% DEA) gradient: 5% to 40% B was maintained for 2.5 min at 40% within 5min, then 5B was maintained for 2.5 min, flow rate: 2.5mL/min, column temperature: 35 ℃"), 97.38% de.
Example 28: synthesis of (3S, 5S,8R,9R,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -13-methylhexahydro-1H-cyclopenta [ a ] phenanthren-3-ol (28)
Figure BDA0003762336660001431
Figure BDA0003762336660001441
1. Pd/C (anhydrous, 200 mg) was added under Ar to a solution of 44 (200mg, 0.480mmol) in MeOH/THF (10 mL/10 mL). The suspension was degassed under vacuum and washed with H 2 The purge was carried out three times. Mixing the mixture in H 2 (50 psi) stirred at 50 ℃ for 48hrs to give a black suspension. The reaction mixture was filtered through a pad of celite and washed with THF (100 mL). The filtrate was concentrated to give solid 28 (30mg, 15%) and solid 82 (30mg, 15%).
28:
1 H NMR(400MHz,CDCl3)δ3.63-3.61(m,1H),1.98-1.76(m,4H),1.72-1.55(m,7H),1.55-1.47(m,4H),1.46-1.23(m,6H),1.22-0.97(m,11H),0.92-0.78(m,12H),0.76-0.54(m,5H)。
LCMS Rt =1.298 min, chromatography over 2 min, 30-90AB, 100% purity, MS ESI C 28 H 47 Calculated value of [ M + H-2H ] 2 O]+383, found 383.
Example 29: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-ethyl-17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (29)
Figure BDA0003762336660001442
Figure BDA0003762336660001451
1. A solution of 1-bromo-3-methylbutane (11.7 g, 78mmol) in THF (8 mL) was added dropwise to Mg (4.35g, 179mmol) and I at 60 deg.C 2 (20 mg) in THF (2 mL). The mixture was stirred at 60 ℃ for 1 hour. The mixture was diluted with THF (10 mL) and used directly. At N 2 Freshly prepared isopentylmagnesium bromide (19.5 mL,3.9M in THF, 76 mmol) was added to a solution of S-200-INT-5E (1.0 g, 2.78mmol) in THF (5 mL) at 0 deg.C. The mixture was stirred at 0 ℃ for 1 hour. Reacting NH 4 Cl (20 mL, saturated aqueous solution) was added to the mixture. The mixture was extracted with EtOAc (2 × 30 mL). The combined organic phases were washed with brine (100 mL) and Na 2 SO 4 Dried, concentrated in vacuo, purified by silica gel (PE/EtOAc =20/1 to 10/1), and purified from CH 3 CN (10 mL) was recrystallized to give 77 (255mg, 21%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.32-5.26(m,1H),3.66-3.59(m,1H),2.42-2.32(m,1H),2.07-1.85(m,4H),1.77-1.58(m,4H),1.55-1.38(m,10H),1.38-1.19(m,5H),1.19-1.00(m,8H),1.00-0.81(m,13H),0.69(s,3H)。
LCMS Rt =1.306 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 29 H 49 O[M+H-H 2 O] + Calculated value 413, found value 413.
2. Pd (OH) under Ar 2 (Anhydrous, 20%,50.0 mg) was added to a solution of 77 (100mg, 232umol) in THF (10 mL) and MeOH (10 mL). The suspension is degassed in vacuo and treated with H 2 The purge was performed three times. Mixing the mixture in H 2 (50 psi) at 50 ℃ for 16 hours. The reaction mixture was filtered through a pad of celite and washed with THF (3 × 10 mL). The filtrate was concentrated. The residue was purified by silica gel chromatography (PE/EtOAc = 20/1) to give 29 (7.00mg, 7%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.68-3.60(m,1H),1.96-1.88(m,2H),1.68-1.60(m,3H),1.53-1.47(m,7H),1.39-1.23(m,13H),1.16-0.95(m,7H),0.90-0.86(m,12H),0.83(s,3H),0.66-0.63(m,4H)。
LCMS Rt =1.603 min, chromatography at 2.0 min, 30-90AB _ELSD, 97% purity, MS ESI C 29 H 49 [M+H-H 2 O] + Calculated value 397 of (a), found value 397.
Example 30: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -3,10, 13-trimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (30)
Figure BDA0003762336660001461
1. At 25 ℃ in N 2 Next, benzoic acid (2.03g, 16.7 mmol) and triphenylphosphine (6.57g, 25.1mmol) were added to a solution of S-500-2-10 (3.5g, 8.39mmol) in THF (30 mL). The mixture was stirred at 25 ℃ for 20mins. At 0 ℃ in N 2 Next, DIAD (5.07g, 25.1mmol) was added. The mixture was stirred at 0 ℃ for 20mins, then warmed to 25 ℃ and stirred for 1h. The reaction mixture was quenched with water (100 mL) and extracted with MTBE (3 × 30 mL). The combined organic phases were washed with brine (60 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude material, which was purified by flash column (0-10% etoac in PE) to give 300mg of crude product S-500-2-9 _1as an oil, which was used directly in the next step.
2._ NaOH (1.14 g in 3mL H 2 O, 28.7 mmol) was added to a solution of S-500-2-9_1 (300mg, 0.576 mmol) in THF (5 mL) and MeOH (3 mL). The mixture was stirred at 50 ℃ for 16 hours. The mixture was washed with saturated NH 4 Cl (20 mL) quenched and extracted with EtOAc (3 × 10 mL). The combined organic phases were washed with brine (30 mL) and Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-10% EtOAc in PE) to give 30 (12mg, 5%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.32-5.28(m,1H),3.63-3.59(m,1H),2.44-2.40(m,1H),2.05-1.90(m,3H),1.80-1.62(m,4H),1.61-1.58(m,3H),1.56-1.30(m,9H),1.28-1.03(m,10H),1.01(s,3H),0.99-0.85(m,10H),0.69(s,3H)。
LCMS t R =1.260 min, 2 min chromatography, 30-90AB _ELSD, purity 100.0%, MS ESI C 28 H 45 [M+H-2H 2 O] + Calculated value 381, found value 381 of (g).
Example 31: synthesis of (1R, 3R, 4S) -4- ((3S, 5S,8R,9S,10S,13S,14S, 17R) -3-hydroxy-10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-17-yl) -1-phenylpentane-1, 3-diol (31)
Figure BDA0003762336660001471
The synthesis of 31 can be seen in example 13.
31:
1 H NMR(400MHz,CDCl 3 )δ7.45-7.28(m,5H),5.02-4.81(m,1H),4.18-3.98(m,1H),3.35(brs,1H),2.47(brs,1H),2.15-1.72(m,8H),1.53-1.31(m,8H),1.30-1.03(m,8H),0.99-0.89(m,4H),0.89-0.78(m,4H),0.75-0.60(m,4H)。
LCMS Rt =1.327 min, chromatography over 2min, 10-80AB _2MIN _E, 100% purity, MS ESI C 31 H 45 F 3 O 3 Na[M+Na] + Calculated 545, found 545.
SFC Rt =1.929 min, IC-3 \ u MeOH (DEA) _40 _u2.5ML, 98.4% de by 10 min chromatography.
Example 32: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-ethyl-17- ((2S, 3S) -3-hydroxy-4-phenylbutan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (32)
Figure BDA0003762336660001481
Figure BDA0003762336660001491
1. t-BuOK (4.64g, 41.4 mmol) was added to Me at 20 ℃ under nitrogen 3 Suspension of SI (8.44g, 41.4 mmol) in dry THF (50 mL). The mixture was stirred at 20 ℃ for 1 hour, and N-005_5 (5g, 13.8mmol) was added. The resulting mixture was warmed to 45 ℃ and stirred for 4 hours. The reaction mixture was cooled to room temperature, quenched with water (50 mL), and extracted with EtOAc (2 × 50 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography on silica gel (0-10% etoac in PE) to give N-005_001 (2.7g, 52%) as a solid.
LCMS Rt =1.324 min, chromatography at 2.0 min, 30-90AB _E, 92% purity, MS ESI C 25 H 41 O[M+H-H 2 O] + Calculated value 357, found value 357.
2. CuI (10.1mg, 0.0534mmol) and PhMgBr (1M in THF, 2.66mL, 2.66mmol) were added to a solution of N-005_001 (200mg, 0.534mmol) in anhydrous THF (20 mL) at 0 deg.C under nitrogen. The mixture was gradually warmed to 15 ℃ and stirred for 16 hours. NH for reaction mixture 4 Aqueous Cl (20 mL) was quenched and extracted with EtOAc (2X 10 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography on silica gel (0-5% EtOAc in PE) to give NA-6-5/6 (190mg, 79%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ7.35-7.27(m,2H),7.25-7.18(m,3H),3.95-3.81(m,1H),2.87-2.39(m,2H),2.04-1.92(m,1H),1.89-1.80(m,1H),1.71-1.58(m,4H),1.56-1.43(m,6H),1.41-1.27(m,5H),1.26-1.18(m,4H),1.18-1.08(m,2H),1.06-0.96(m,5H),0.92-0.79(m,8H),0.73-0.55(m,4H)。
N-6-5/6 (190mg, 0.420mmol) was separated by prep.HPLC (column: YMC-Actus Triart C18 100 x 30mm x 5um; conditions: water (0.05% HCl) -ACN; gradient: 90-100% B; flow rate: 25 mL/min) to give solid 93 (56mg, 30%) and solid 32 (12mg, 6%).
32:
1 H NMR(400MHz,CDCl 3 )δ7.36-7.29(m,2H),7.25-7.19(m,3H),3.89-3.83(m,1H),2.79-2.72(m,1H),2.49-2.40(m,1H),2.05-1.98(m,1H),1.92-1.79(m,2H),1.72-1.51(m,9H),1.44-1.31(m,5H),1.30-1.09(m,7H),1.08-0.96(m,5H),0.92-0.81(m,7H),0.74-0.63(m,4H)。
LCMS Rt =1.343 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 31 H 47 O[M+H-H 2 O] + Calculated 435, found 435.
Example 33: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-4- (3-methyloxetan-3-yl) but-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (33)
Figure BDA0003762336660001501
1. To a solution of N-014-005-uu 1 (10g, 97.9mmol) in DCM (100 mL) at 25 ℃ was added 1-methyl-1H-imidazole (16.0g, 195mmol) and TEA (19.7g, 195mmol). TsCl (37.1g, 195mmol) was added to the solution. The reaction mixture was stirred at 25 ℃ for 2 hours. The mixture was washed with water (2X 100 mL), brine (100 mL), na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-014-005_2 (25 g, crude) as a pale yellow solid, which was purified by column chromatography on silica gel (0-15% etoac in PE) to afford N-014-005_2 (23.6 g, 95%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ7.80-7.68(m,2H),7.41-7.26(m,2H),3.40-3.29(m,4H),4.12-4.00(s,2H),2.44(s,3H),1.28(s,3H)。
2. LiBr (13.5g, 156mmol) was added to a solution of N-014-005 (u 2 (10g, 39.0 mmol) in acetone (100 mL). The mixture was stirred at 65 ℃ for 1 hour. The mixture was quenched with water (200 mL) at 0 ℃ and hexanes (3X 200 mL)) And (4) extracting. The combined organic phases were washed with brine (50 mL) and Na 2 SO 4 Dried, filtered and concentrated to give N-014-005 (2.54 g, crude) as a yellow liquid.
1 H NMR(400MHz,CDCl 3 )δ4.50-4.30(m,4H),3.64(s,2H),1.58(s,1H),1.43(s,3H)。
Figure BDA0003762336660001511
3. In N 2 Adding Mg (807mg, 33.2mmol) and I at 50-55 deg.C 2 (1 mg) to a suspension in THF (2 mL) was added dropwise a solution of N-014-005_3 (2.5g, 15.1mmol) in THF (8 mL). The mixture was stirred at 55 ℃ for 1h. The mixture was diluted with THF (10 mL) and used directly in the next step without monitoring. To a solution of N-14-12 (1.01g, 2.83mmol) in THF (10 mL) at 0 deg.C was added the freshly prepared 3- [ (bromomagnesium (broomagnesio)) methyl group ]-3-methyloxetane (15 mmol in 20mL THF). The mixture was stirred at 15 ℃ for 4h. Adding NH to the mixture 4 Cl (20mL, 10. Aq.). The mixture was extracted with EtOAc (30 mL). The organic layer was separated and concentrated in vacuo. The residue was purified by flash column (0-30% EtOAc in PE) to give a mixture (190mg, 15%) as a white solid, which was purified by SFC (column: AD (250mm. Multidot. 30mm,5 um), conditions: 0.1% NH3H2O ETOH, gradient: 50% to 50%, flow rate (mL/min): 60mL/min,25 ℃) to give 33 (peak 1, 110mg, 9%) and 70 (peak 2, 30mg, impure) as white solids. Impure 70 (30 mg, impure) was purified by column chromatography on silica gel (15% etoac in PE) to give 70 (10 mg, 5%) as a white solid.
33:
1 H NMR(400MHz,CDCl 3 )δ5.30-5.26(m,1H),4.59-4.70(m,1H),4.50-4.48(m,1H),4.36-4.33(m,1H),3.83(s,1H),2.40-2.33(m,1H),2.10-1.50(m,17H),1.49-1.35(m,9H),1.30-0.80(m,13H),0.68(s,3H)。
LCMS Rt =1.069 min, 3 min chromatography, 30-90AB _2MIN _E.M, 100% purity, MS ESI C 29 H 49 O 3 [M+H] + Calculated value 445, found value 445.
Example 34: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-ethyl-17- ((2S, 3R) -3-hydroxy-4-methylpentan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (34)
Figure BDA0003762336660001521
Figure BDA0003762336660001531
1. N-8-7_ (500mg, 1.38mmol) in THF (5 mL) was added to a solution of isopropyl magnesium chloride (708mg, 3.44mL,2M in THF) in THF (5 mL) at 0 ℃. The mixture was stirred at 25 ℃ for 4 hours. Adding NH to the mixture 4 Cl (20mL, 10. Aq.). The mixture was extracted with EtOAc (2 × 30 mL). The organic layer was separated and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography eluting with PE/EtOAc =3/1 to give N-8-11_1 (170mg, 30%) as a solid. The impure product (120 mg) was further purified by silica gel chromatography eluting with PE/EtOAc =3/1 to give N-8-11_1 (50mg, 42%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.20-3.10(m,1H),2.00-1.80(m,2H),1.70-1.60(m,2H),1.55-1.40(m,4H),1.39-0.95(m,19H),0.90-0.80(m,15H),0.70-0.60(m,5H)。
2. DMP (457mg, 1.08mmol) was added to a solution of N-8-11 \u1 (220mg, 0.543mmol) in DCM (5 mL). After stirring for 10 min at 25 ℃ the reaction mixture was saturated with NaHCO 3 The aqueous solution (50 mL) was quenched until the pH of the aqueous layer became about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (100 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3X 100 mL), saturated NaHCO 3 (100 mL), brine (40 mL), na 2 SO 4 Dried, filtered and concentrated to give crude N-8-11 \u2 (140mg, 64%) asAnd (3) a solid.
LCMS Rt =1.300 min, chromatography at 2.0 min, 30-90AB (u 2MIN) E, 100% purity, MS ESI C 27 H 45 O[M+H-H 2 O] + The calculated value 385 of (1), the measured value 385.
3.NaBH 4 (1.17g, 17.3mmol) was added five times every 5 minutes to a solution of N-8-11/-2 (140mg, 0.347mmol) in MeOH (2 mL) and THF (2 mL). The mixture was stirred at 15 ℃ for 30 minutes. Mixing the mixture with sat 4 Cl (50 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases are treated with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (25% etoac in PE) to give N-8-11_1 (140 mg, impure). N-8-11 \/1 was purified by Combi-flash (25% EtOAc in PE) to yield solid 34 (50 mg, impure) and solid 65 (10 mg, impure).
N-8-11-u 1 (50mg, 0.123mmol, impure) was purified by combi-flash (25% EtOAc in PE) to give 34 (30 mg, impure) as a solid.
N-8-11 \ (30mg, 0.0741mmol, impure) was purified by Combi-flash (25% EtOAc in PE) to give 34 (9mg, 30%) as a solid.
34:
1 H NMR(400MHz,CDCl 3 )δ3.18-3.07(m,1H),1.98-1.81(m,2H),1.71-1.58(m,6H),1.53-1.31(m,7H),1.30-0.98(m,14H),0.97-0.78(m,14H),0.70-0.60(m,4H)。
LCMS Rt =4.387 min, chromatography over 7.0 min, 30-90AB u 7MIN u E, 97.6% purity, MS ESI C 27 H 45 [M+H-2H 2 O] + Found 369, found 369.
Example 35: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -3- (methoxymethyl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (35)
Figure BDA0003762336660001541
Figure BDA0003762336660001551
1. NaOH (71.9mg, 180mmol) was added to a solution of N-4-4B (20mg, 0.0361mmol) in THF/MeOH (2 mL) at 25 ℃. The reaction mixture was warmed to 50 ℃ and stirred for 1h. The reaction mixture was cooled and water (20 mg) was added. The aqueous phase was extracted with EtOAc (3 × 10 mL). The combined organic phases were washed with saturated brine (2 × 20 mL) and anhydrous Na 2 SO 4 Dry, filter and concentrate. The residue was purified by flash column (0-30% EtOAc in PE) to provide 35 (8 mg, 50%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ3.63-3.61(m,1H),3.41-3.38(m,5H);2.51(s,1H);1.97-1.81(m,1H),1.71-1.54(m,8H),1.51-1.48(m,4H),1.25-1.10(m,15H),0.99-0.80(m,9H),0.78-0.75(m,4H),0.71-0.59(m,4H)。
LCMS Rt =1.301 min, chromatography at 2.0 min, 30-90AB, 96% purity, MS ESI C 29 H 48 O[M+H-2H 2 O] + Calculated value 413, found value 413.
Example 36: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -3,10, 13-trimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (36)
Figure BDA0003762336660001552
Figure BDA0003762336660001561
1. Pd (OH) 2 (200 mg) was added to a solution of 30 (100mg, 0.239mmol) in MeOH (10 mL). The mixture was heated at 50 ℃ in H 2 Stirring was performed under (50 Psi). The mixture was filtered, concentrated and purified by combi-flash (0-10% etoac in PE) to give 47 (21mg, 21%) and 36 (1mg, 1%) as solids.
37:
1 H NMR(400MHz,CDCl 3 )δ3.66-3.55(m,1H),2.05-1.77(m,3H),1.72-1.63(m,3H),1.55-1.48(m,3H),1.47-1.31(m,9H),1.29-1.12(m,13H),1.11-1.00(m,3H),0.96(s,3H),0.93-0.87(m,9H),0.67(s,3H)。
LCMS t R =1.296 min, chromatography over 2 min, 30-90ab _elsd, purity 100.0%, MS ESI C 28 H 47 [M+H-2H 2 O] + Calculated value 383 of (d), found value 383.
Example 37: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-4-methylpentan-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (37)
Figure BDA0003762336660001562
Figure BDA0003762336660001571
N-014-001_1 (150mg, 0.37mmol) was purified by SFC (column: chiralpak AS-H250: 30 5u; conditions: 0.1% NH 3 H 2 O EtOH; and starting B:20 percent; and B, ending: 20 percent; flow rate (ml/min): 65 To give 37 (peak 2, 46mg, 31%) and 27 (peak 1, 27mg, 18%) as solids.
014-001A:
1 H NMR(400MHz,CDCl 3 )δ5.35-5.28(m,1H),3.18-3.09(m,1H),2.39-2.35(m,1H),2.06-1.81(m,4H),1.73-1.57(m,6H),1.54-1.41(m,8H),1.40-1.26(m,3H),1.24-1.11(m,3H),1.10-0.97(m,6H),0.96-0.92(m,1H),0.90-0.85(m,5H),0.84-0.76(m,4H),0.69(s,3H)。
LCMS Rt =1.207 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, MS ESI C 27 H 45 O[M+H-H 2 O] + The calculated value 385 of (1), the measured value 385.
SFC Rt =2.635 min, 10 min chromatography, OJ _ 3u EtOH _DEA5 _, 40_, 25ML ("column: chiralcel OJ-3X 4.6mm I.D.,3um mobile phase: A: CO 2B: ethanol (0.05% DEA) gradient: 5% to 40% B was maintained for 2.5 min at 40%, then 5% B was maintained for 2.5 min, flow rate: 2.5mL/min, column temperature: 35 ℃"), 98.66 de.
Example 38: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-ethyl-17- ((2S, 3S) -3-hydroxyhex-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (38)
Figure BDA0003762336660001572
Figure BDA0003762336660001581
1. Propylmagnesium bromide (3.34mL, 6.69mmol,2M in THF) was slowly added to a solution of S-500-6-1 \1 (800mg, 2.23mmol) in THF (30 mL) at 0 deg.C. After the addition, the mixture was stirred at 15 ℃ for 1 hour. The mixture was washed with saturated NH 4 Cl (40 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases were washed with brine (2 × 30 mL) and Na 2 SO 4 Dried, filtered and concentrated and purified by combi-flash (0-15% EtOAc in PE) to give 72 (500mg, 56%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.72-3.64(m,1H),2.41-2.31(m,1H),2.07-1.85(m,4H),1.77-1.69(m,1H),1.62-1.54(m,3H),1.52-1.38(m,9H),1.37-1.16(m,6H),1.15-1.01(m,7H),0.99-0.88(m,7H),0.87-0.82(m,3H),0.68(s,3H)。
LCMS Rt =4.979 min, chromatography at 7.0 min, 30-90AB _E, 98.8% purity, MS ESI C 27 H 43 [M+H-2H 2 O] + The calculated value 367 and the actual value 367 of (a).
2. Pd (OH) 2 (300 mg, anhydrous) was added to a solution of 72 (150mg, 0.372mmol) in MeOH (20 mL). The mixture was heated at 50 ℃ in H 2 Stirring was carried out under (50 Psi)For 48 hours. The mixture was filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 23 (9mg, 6%) and 38 (43mg, 29%) as solids.
38:
1 H NMR(400MHz,CDCl 3 )δ3.71-3.62(m,1H),1.99-1.82(m,2H),1.70-1.56(m,6H),1.54-1.45(m,3H),1.44-1.38(m,3H),1.37-1.17(m,10H),1.16-1.01(m,5H),1.00-0.85(m,11H),0.82(s,3H),0.70-0.60(m,4H)。
LCMS Rt =1.397 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 27 H 45 [M+H-2H 2 O] + Found 369, found 369.
Example 39: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3- (methoxymethyl) -10, 13-dimethyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybut-2-yl) -2,3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (39)
Figure BDA0003762336660001591
1. In N 2 Next, t-BuOH (600 mL) was added to a three-necked round-bottom flask at 35 ℃ followed by t-BuOK (101g, 905mmol). After stirring for 30mins at 35 ℃ N-004-029 (50g, 151mmol) was added to the above mixture and stirred for 1 hour at 35 ℃. The reaction mixture was poured into 5% aqueous acetic acid (2L) during which time the temperature was kept below 10 ℃. Ice-water (1L) was added. The pH of the mixture was determined using NaHCO 3 Adjusted to about 7-8 and filtered. The filter cake was dissolved in DCM (1.5L). The combined organic phases were washed with water (2X 500 ml), brine (2X 500 ml) and Na 2 SO 4 Dried, filtered and concentrated in vacuo at below 35 ℃ to give N-004-029 (45 g, crude) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.35-5.32(m,1H),3.71-3.58(m,1H),3.38-3.25(m,2H),2.90-2.78(m,1H),2.55-2.20(m,2H),2.13-1.92(m,3H),1.90-1.59(m,5H),1.46-1.14(m,10H),1.12-0.96(m,6H),0.72(s,3H)。
2. At 0 ℃ in N 2 n-BuLi (108mL, 272mmol,2.5M in n-hexane) was added dropwise to Me 3 SI (73.8g, 362mmol) in dry THF (300 mL). After stirring at 0 ℃ for 30mins, a solution of N-004-029 (30g, 90.7 mmol) in anhydrous THF (600 mL) was added at-40 ℃. The mixture was stirred at-40 ℃ for 2 hours and at 25 ℃ for 16 hours. The reaction mixture was poured into ice-water (1L). The aqueous phase was extracted with EtOAc (2 × 500 mL). The combined organic phases were washed with brine (2X 500 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated, and the residue purified by flash column (0-20% EtOAc in PE) to give N-004-029_3 (1.8g, 6%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.33-5.25(m,1H),3.66-3.61(m,1H),3.39-3.31(m,1H),2.93-2.86(m,1H),2.59-2.53(m,1H),2.20-1.93(m,4H),1.89-1.14(m,15H),1.12-0.90(m,9H),0.71(s,3H)。
3. At 25 ℃ in N 2 MeONa (5.61g, 104mmol) was then added to a solution of N-004-029_3 (1.8g, 5.22mmol) in MeOH (20 mL). After stirring at 50 ℃ for 12hrs, water (100 mL) was added to the mixture and stirred for 10mins. The aqueous phase was extracted with EtOAc (2 × 80 mL). The combined organic phases were washed with saturated brine (2 × 50 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-30% EtOAc in PE) to give N-004-029_4 (1.5g, 76%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.31-5.28(m,1H),3.70-3.54(m,1H),3.40-3.35(m,6H),3.28-3.16(m,2H),2.40-2.35(m,1H),2.09-1.90(m,5H),1.87-1.57(m,11H),1.34-1.06(m,10H),0.70(s,3H)。
4. DMP (2.53g, 5.97mmol) was added to a solution of N-004-029_4 (1.5 g, 3.98mmol) in DCM (30 mL) at 25 ℃ and after stirring for 30 minutes at 25 ℃. The reaction mixture was quenched with saturated NaHCO at 25 deg.C 3 Quench (50 mL). DCM (50 mL) was added to the mixture and stirred for 10 min. The DCM phase was separated and saturated Na was used 2 S 2 O 3 Aqueous solution (2X 50 mL), brine (2X 50 mL), washed with Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (5-25% EtOAc in PE) to give N-004-029_5 (0.6 g, 40%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.59-9.57(m,1H),5.32-5.29(m,1H),3.37(s,3H),3.30-3.15(m,2H),2.44-2.31(m,2H),2.13-1.40(m,16H),1.27-1.02(m,10H),0.73(s,3H)。
5. CsF (607mg, 4.00mmol) was added to a solution of N-004-029-5 (0.6g, 1.60mmol) in anhydrous THF (20 mL) at 0 deg.C. After stirring for 20 minutes, TMSCF was added at 0 deg.C 3 (568mg, 4.00mmol) and the mixture was stirred for 1 hour. Reacting TBAF.3H 2 O (2.02g, 6.40mmol) was added to the mixture, which was stirred at 50 ℃ for 1 hour. The reaction mixture was poured into ice-water (50 mL). The aqueous phase was extracted with EtOAc (2 × 80 mL). The combined organic phases were washed with saturated brine (2X 80 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-30% EtOAc in PE) to give N-004-029A (450mg, 63%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.33-5.29(m,1H),4.11-3.99(m,1H),3.37(s,3H),3.30-3.19(m,2H),2.54(s,1H),2.43-2.36(m,1H),2.26-1.82(m,7H),1.78-1.61(m,5H),1.34-0.80(m,15H),0.75-0.67(m,3H)。
N-004-029A (0.45g, 1.01mmol) was purified by SFC (column: AD (250mm. 30mm,5 um), conditions: 0.1% NH 3 H 2 O ETOH, start B:30%, end B: 30%) to give white solid 39 (PK 1:120mg, 26.7%) and white solid 95 (PK 2:200mg, 44.5%).
39 was confirmed by NOE.
39:
1 H NMR(400MHz,CDCl 3 )δ5.32-5.29(m,1H),4.06-3.99(m,1H),3.37(s,3H),3.30-3.19(m,2H),2.54(s,1H),2.43-2.36(m,1H),2.25-2.19(m,1H),2.15-2.07(m,1H),2.04-1.60(m,9H),1.55-1.34(m,5H),1.25-0.88(m,11H),0.70(s,3H)。
LCMS Rt =1.078 min, chromatography over 2min, 30-90AB (u 2MIN) (100% purity) MS ESI C 24 H 34 F 3 O[M-CH 5 O 2 ] + Calculated 395, found 395.
Example 40: synthesis of (3S, 5S,8R,9R,10S,13S,14S, 17R) -3-ethyl-17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -13-methylhexahydro-1H-cyclopenta [ a ] phenanthren-3-ol (40)
Figure BDA0003762336660001621
1. Pd (OH) at 25 ℃ under Ar 2 (100 mg, anhydrous) was added to a solution of 3 (30mg, 0.072mmol) in MeOH/THF (5 mL/5 mL). The reaction was carried out at 50 ℃ in H 2 Stirring was carried out for 48h next (50 Psi). The mixture was filtered and the filtrate was concentrated in vacuo to give the crude product, which was purified by silica gel column (PE/EtOAc = 10/1-5/1) to give 41 (10mg, 33%) as a solid.
1 H NMR(400MHz,CDCl3)δ3.62-3.59(m,1H),1.99-1.91(m,1H),1.88-1.76(m,2H),1.74-1.61(m,6H),1.46-1.29(m,6H),1.26-1.09(m,9H),1.08-0.99(m,6H),0.95-0.78(m,14H),0.74-0.58(m,5H)。
LCMS Rt =1.491 min, 2min chromatography, 30-90AB, 99% purity, MS ESI C 28 H 47 Calculated value of [ M + H-2H ] 2 O] + 383, found 383.
Example 41: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxyhept-5-yn-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (41)
Figure BDA0003762336660001622
Figure BDA0003762336660001631
1. n-BuLi (13.8 m) was added at-20 deg.CL,34.5mmol,2.5m in hexanes) was added dropwise to a solution of but-2-yne (1.86g, 34.5mmol) in THF (100 mL). The solution was stirred at-20 ℃ for 2.5hrs, then cooled to-78 ℃ and N-8-7_1 (5.0 g,13.8 mmol) in THF (100 mL) was added. The solution was stirred at this temperature for 30mins, then-20 ℃ for 1 hour, then 20 ℃ for 18 hours. The resulting gel was poured into saturated NH 4 Cl (100 mL) was quenched and then extracted with EtOAc (2X 150 mL). The combined organic layers were washed with water (40 mL) and brine (40 mL), and Na 2 SO 4 Dried, filtered and eluted with 0-20% EtOAc in PE by flash column purification to give oil N-8-7 \u2B (1 g, crude) and oil N-8-7 \u2B (1.7 g, crude).
1 H NMR(400MHz,CDCl 3 )δ3.90-3.75(m,1H),2.45-2.35(m,1H),2.25-2.05(m,3H),1.95-1.85(m,2H),1.85-1.40(m,8H),1.40-1.20(m,9H),1.20-0.75(m,18H),0.65(s,4H)。
SFC peak 1: rt =3.008 min, 10 min chromatography, AD _3 \ u EtOH _ \ DEA _5_40_25ML ("column: chiralpak AD-3X 4.6mm I.D.,3um mobile phase: A: CO 2B: ethanol (0.05%/DEA) gradient: 5% to 40% B in 5 min and 2.5 min at 40%, then 5B for 2.5 min, flow rate: 2.5mL/min, column temperature: 35 ℃).
2. The crude N-8-7_2B (250mg, 0.868mmol) was further purified by SFC (column: AD (250mm. 30mm, 10um)), gradient: 35-35% B (A =0.1% NH) 3 /H 2 O, B = EtOH), flow rate: 60 mL/min) to give 41 as a solid (peak 2, 81mg, 33%) and 68 as a solid (peak 1, 78mg, 31%).
41:
1 H NMR(400MHz,CDCl 3 )δ3.82-3.70(m,1H),2.79-2.08(m,2H),2.00-1.90(m,1H),1.80(s,4H),1.78-1.69(m,1H),1.69-1.42(m,10H),1.40-1.31(m,4H),1.31-1.18(m,4H),1.18-0.92(m,6H),0.92-0.85(m,7H),0.82(s,3H),0.66(s,3H)。
LCMS Rt =1.206 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, calculated value C of MS ESI 28 H 45 O[M+H-H 2 O] + 397Found 397.
SFC Rt =5.823 min, AD _3_EtOH _DEA5 _40 _25MLin 10 min chromatography, 100% de.
Example 42: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (42)
Figure BDA0003762336660001641
Figure BDA0003762336660001651
1. 4-Methylbenzenesulfonic acid (2.70g, 15.7 mmol) was added to a solution of N-4-1/-1 (50g, 157mmol) in MeOH (500 mL) at 25 ℃. The mixture was stirred at 65 ℃ for 1 hour. The reaction mixture was cooled to 25 ℃ and TEA (2.16mL, 15.7mmol) was added. The mixture was stirred for 0.5h. The precipitate was collected by filtration and washed with methanol (2 × 100 mL) to give N-4-1_2 (50 g, crude) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.25-3.05(m,6H),2.60-2.40(m,1H),2.20-2.05(m,4H),2.00-1.95(m,1H),1.90-1.80(m,1H),1.75-1.50(m,6H),1.49-1.05(m,12H),1.04-0.95(m,1H),0.78(s,3H),0.59(s,3H)。
2. t-BuOK (23.0 g, 205mmol) was added to a solution of bromo (methyl) triphenyl phosphorane (73.2 g, 205mmol) in THF (500 mL) at 25 ℃. The mixture was heated to 45 ℃ and stirred for 1 hour. N-4-1_2 (50g, 137mmol) was added. The mixture was stirred at 45 ℃ for 2 hours. Subjecting the mixture to NH 4 Cl (200 mL) quenched and extracted with THF (3 × 100 mL). The organic layer was washed with brine (200 mL) and Na 2 SO 4 Dried and filtered to give a mixture (50g, 500ml) which was used in the next step without further purification.
3. Aqueous HCl (207mL, 1M in water) was added to a solution of N-4-1_3 (50g, 138mmol) in THF (500 mL). The mixture was stirred at 25 ℃ for 0.5h. Mixing the componentsThe material was filtered and the filter cake was dissolved in DCM (200 mL) and washed with brine (100 mL), anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-4-1 \u4 (39g, 90%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.70(s,1H),2.45-2.20(m,3H),2.15-2.00(m,3H),1.90-1.65(m,8H),1.60-1.50(m,2H),1.45-1.05(m,8H),1.00(s,3H)0.90-0.85(m,1H),0.80-0.75(m,1H),0.58(s,3H)。
4. CsF (25.9g, 171mmol) and TMSCF 3 (24.3g, 171mmol) is added to a solution of N-4-1_4 (27g, 85.8mmol) in THF (200 mL). The mixture was stirred at 10 ℃ for 1 hour. Water (10 mL) and TBAF.3H 2 O (30 g) was added to the mixture. The mixture was stirred at 30 ℃ for a further 2 hours. The mixture was concentrated in vacuo. The residue was dissolved in EtOAc (500 mL), washed with water (2X 500 mL), and washed with Na 2 SO 4 Dried, filtered, concentrated in vacuo and purified by flash column (DCM/EtOAc (1) in PE, 0-10%) to give N-4-1_5 (27g, 82%) and N-4-1_5a (3.5g, 11%) as a solid.
N-4-1_5:
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.70(s,1H),2.12-1.94(m,3H),1.89-1.78(m,2H),1.75(s,3H),1.72-1.60(m,5H),1.58-1.48(m,2H),1.45-1.09(m,10H),1.01-0.89(m,1H),0.85(s,3H),0.78-0.68(m,1H),0.56(s,3H)。
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.70(s,1H),2.09-1.99(m,1H),1.89-1.78(m,2H),1.75(s,3H),1.72-1.52(m,9H),1.45-1.06(m,10H),1.00-1.81(m,2H),0.79(s,3H),0.56(s,3H)。
5. 9-BBN dimer (29g, 119mmol) was added to a solution of N-4-1 \u5 (23g, 59.8mmol) in THF (250 mL) and the mixture was heated at 40 ℃ in N 2 Stirred for 16 hours. Ethanol (34.3ml, 598 mmol) and NaOH (119ml, 5m,598 mmol) were added to the reaction mixture. The mixture became clear. H is added dropwise at 25 DEG C 2 O 2 (59.8mL, 10M, 598mmol) and the internal temperature was raised to reflux (70 ℃ C.). After addition the mixture was cooled to 30 ℃. To the mixture was added Na 2 SO 3 (100mL, 20. Though aq.). The organic layer was separated and poured into water (800 mL). A solid was formed. The mixture was filtered and the solid was washed with water, dried in vacuo and triturated with MeCN (250 mL) to give a solid. The solid was triturated from MeOH/water (250 mL/12.5 mL) at 60 ℃ and filtered after cooling to 15 ℃. The solid was dried under vacuum to give N-4-1_6 (16.4g, 68%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.69-3.60(m,1H),3.39-3.29(m,1H),2.09-2.01(m,1H),1.99-1.92(m,1H),1.87-1.75(m,2H),1.72-1.43(m,7H),1.42-1.07(m,11H),1.03(d,J=6.8Hz,3H),1.01-0.86(m,3H),0.85(s,3H),0.73-0.69(m,1H),0.67(s,3H)。
6. Water (223mg, 12.4 mmol) and DMP (10.5g, 24.8mmol) were added to a suspension of N-4-1_6 (5g, 12.4 mmol) in DCM (200 mL). The mixture was stirred at 15 ℃ for 15 minutes. The mixture is washed with NaHCO 3 /Na 2 S 2 O 3 (200 mL/200mL, sat.) twice, over Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-4-1_7 (4.5g, 90%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.60-9.51(m,1H),2.40-2.30(m,1H),2.12-1.78(m,5H),1.75-1.59(m,4H),1.57-1.15(m,11H),1.14-0.84(m,8H),0.78-0.63(m,5H)。
7. In N 2 A solution of 1-bromo-3-methylbutane (2.79g, 18.5 mmol) in THF (8 mL) was added dropwise to Mg (899mg, 37mmol) and I at 50-55 deg.C 2 (1 mg) suspension in THF (2 mL). The mixture was stirred at 55 ℃ for 1hr to obtain an isopentyl magnesium bromide solution. Freshly prepared isopentyl magnesium bromide (18.5 mmol in 10mL THF) was added to a solution of N-4-1/-7 (0.5g, 1.24mmol) in THF (5 mL) at 0 deg.C. The mixture was stirred at 15 ℃ for 2 hours. Adding NH to the mixture 4 Cl (20mL, 10. Aq.). The mixture was extracted with EtOAc (2 × 30 mL). The combined organic layers were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give N-4-2 (0.6 g, crude) as a solid.
8. Water (1 drop) and DMP (1.06g, 2.52mmol) were added to a solution of N-4-2 (0.6g, 1.26mmol) in DCM (20 mL) at 15 ℃. The mixture was stirred at 15 ℃ for 1h. The mixture is washed with NaHCO 3 /Na 2 S 2 O 3 (20 mL/20mL, sat.) was washed twice with Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-4-1 \u8 (0.6 g, crude) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.59-2.30(m,3H),2.11-1.78(m,4H),1.75-1.36(m,13H),1.35-0.98(m,11H),0.91-0.82(m,10H),0.78-0.70(m,1H),0.67(s,3H)。
9. NaBH is reacted at 15 ℃ 4 (0.96g, 25.4 mmol) was added portionwise to a solution of N-4-1_8 (0.6g, 1.27mmol) in THF (10 mL) and MeOH (5 mL). The mixture was stirred at 15 ℃ for 30mins. Adding NH to the mixture 4 Cl (50mL, 10%). The mixture was extracted with EtOAc (2 × 50 mL). The combined organic layers were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo and purified by flash column (0-15% etoac in PE) to give impure 42 and 85. N42 was triturated from MeCN (10 mL) at 15 ℃ and dried in vacuo to give 42 (153mg, 25%) as a solid. 85 was purified by flash column (0-15% EtOAc in PE) to give an oil, which was treated with MeCN (5 mL) and water (5 mL), and concentrated in vacuo to give 85 (70mg, 12%) as a solid.
42:
1 H NMR(400MHz,CDCl 3 )δ3.66-3.55(m,1H),2.01-1.78(m,6H),1.71-1.59(m,4H),1.51-1.15(m,16H),1.09-1.02(m,3H),0.92-0.81(m,13H),0.72-0.61(m,4H)。
LCMS Rt =1.378 min, chromatography at 2.0 min, 30-90_AB _E, purity 100%, MS ESI C 28 H 46 F 3 O[M+H-H 2 O] + Calculated value 455 of (a), measured value 455.
HPLC Rt =5.38 min, chromatography at 10.0 min, 50-100_ab \ue, purity 99.58%.
Example 43: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3-Ethyl-10, 13-dimethyl-17- ((2S, 3R) -4, 4-trifluoro-3-hydroxybutan-2-yl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (43)
Figure BDA0003762336660001691
1. To a solution of 62 (160mg, 0.373mmol) in MeOH (2 mL) and THF (1 mL) was added Pd (OH) 2 (0.2g,<1% water). The solution was hydrogenated under 50psi of hydrogen at 50 deg.C for 16 hours. The mixture was then filtered through a pad of celite and the filtrate was concentrated in vacuo. The residue was purified by flash column (PE/EtOAc =10/1 to 5/1) to give 43 (27mg, 17%) and 16 (117mg, 73%) as white solids.
43:
1 H NMR(400MHz,CDCl3)δ.4.05-3.99(m,1H),1.99-1.81(m,5H),1.79-1.72(m,1H),1.70-1.56(m,3H),1.53-1.35(m,7H),1.35-1.07(m,12H),1.04-1.02(m,3H),0.97(s,3H),0.92(t,J=8Hz,3H),0.70(s,3H)。
LCMS Rt =1.271 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 25 H 40 F 3 Calculated value of O [ M + H-H ] 2 O] + 413, found value 413.
Example 44: synthesis of (3S, 8R,9S,10R,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -13-methyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (44)
Figure BDA0003762336660001692
Figure BDA0003762336660001701
1. A solution of 1-bromo-3-methylbutane (4 g, 26.4mmol) in THF (27 mL) was added dropwise to a suspension of Mg (947mg, 39.5mmol) and I2 (33.5mg, 0.132mmol) in THF (3 mL) at 60 ℃. The mixture was stirred at 60 ℃ for 1 hour. In N 2 At 0 DEG CFreshly prepared isopentyl magnesium bromide (30mL, 0.88M in THF, 26.4 mmol) was added to a solution of S-500-15-2 \u1 (800mg, 2.32mmol) in THF (2 mL). The mixture was stirred at 0 ℃ for 1h. To the mixture was added NH4Cl (50 mL, saturated aqueous solution) and the mixture was extracted with EtOAc (2 × 50 mL). The combined organic phases were washed with brine (100 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product, which was purified by silica gel (PE/EtOAc =10/1 to 5/1) to give 44 (720mg, 75%) as a white solid.
1H NMR(400MHz,CDCl3)δ5.40-5.38(m,1H),3.63-3.61(m,1H),2.23-2.21(m,1H),2.10-1.74(m,7H),1.69-1.58(m,2H),1.54-1.34(m,8H),1.33-1.00(m,11H),0.95-0.75(m,14H),0.70(s,3H)。
LCMS Rt =1.289 min, chromatography over 2 min, 30-90AB, 100% purity, MS ESI C28H45 calculated [ M + H-2H2O ] +381, found 381.
Example 45: synthesis of (3S, 5R,8R,9R,10S,13S,14S, 17R) -3- (methoxymethyl) -13-methyl-17- ((2S, 3R) -4, 4-trifluoro-3-hydroxybutan-2-yl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (45)
Figure BDA0003762336660001702
Figure BDA0003762336660001711
1. TsOH (6.26g, 36.4 mmol) was added to a solution of G-020-004 \u1 (100g, 364mmol) in dry methanol (1L). The mixture was stirred at 60 ℃ for 18 hours. The reaction mixture was concentrated to remove most of the solvent, and treated with Et 3 N (3.7 g) was neutralized, diluted with EtOAc (600 mL), washed with water (500 mL) and brine (500 mL). The organic layer was concentrated to give G-020-004 u 2 (133G, crude) as an oil.
1 H NMR(400MHz,CDCl 3 )δ3.20(s,3H),3.14(s,3H),2.63-2.39(m,2H),2.14-2.03(m,2H),1.97-1.89(m,2H),1.86-1.77(m,3H),1.64-1.60(m,2H),1.56-1.49(m,3H),1.47-1.42(m,2H),1.40-1.32(m,2H),1.29-1.23(m,3H),1.16-1.06(m,2H),0.87(s,3H)。
2. Under nitrogen at 20 ℃ to Ph 3 To a suspension of PEtBr (308g, 830mmol) in anhydrous THF (700 mL) was added t-BuOK (93.1g, 830mmol). After stirring at 20 ℃ for 1 hour, a solution of G-020-004 u 2 (133g, 415mmol) in anhydrous THF (300 mL) was added to the mixture. The resulting mixture was warmed to 50 ℃ and stirred for 4 hours. The reaction mixture was cooled, and water (400 mL) and saturated NH were used 4 Cl (300 mL) and stirred for 30 min. The organic layer was separated and the aqueous phase was extracted with THF (300 mL). The combined organic layers were used directly in the next step.
3. To a solution of G-020-004 (137g, 412mmol, theory) in THF (1.3L) was added aqueous HCl (1M, 618mL, 618mmol). After stirring for 1 hour at 20 ℃ the reaction mixture was taken up with saturated NaHCO 3 Quenched (800 mL) and extracted with EtOAc (2 × 500 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to give a solid (300 g). The solid was triturated from petroleum ether (800 mL) for 18 hours. The solid was filtered off and the filter cake was washed with petroleum ether (400 mL). The filtrate was concentrated to give a residue (117 g). The residue was purified by column chromatography on silica gel (0-10% EtOAc in PE) to give N-004-023_5 (70 g) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.17-5.09(m,1H),2.65-2.55(m,1H),2.43-2.34(m,1H),2.33-2.15(m,6H),2.11-2.05(m,1H),1.83-1.70(m,2H),1.68-1.64(m,4H),1.63-1.59(m,2H),1.58-1.46(m,3H),1.42-1.25(m,3H),1.25-1.14(m,3H),0.91(s,3H)。
4. A stirred solution of trimethyliodothionium oxide (30.5 g, 139mmol) and t-BuOK (15.5 g, 139mmol) in DMSO (200 mL) was heated at 60 ℃ in N 2 Heating for 1.0h; n-004-023 u 5 (20g, 69.8mmol) was added to the reaction mixture and stirred at 60 ℃ for 10mins. The reaction was treated with water (1000 mL). The aqueous phase was extracted with EtOAc (2 × 500 mL). The combined organic phases were washed with water (2X 500 mL), brine (300 mL), and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give N-004-023 u 6 (20.5 g, crudeBiomass) which is a solid.
1 H NMR(400MHz,CDCl 3 )δ5.13-5.10(m,1H),2.62-2.60(m,2H),2.25-2.20(m,5H),2.00-1.48(m,12H),1.46-1.00(m,8H),0.98-0.89(m,4H)。
5. At 25 ℃ in N 2 MeONa (18.4g, 341mmol) was then added to a solution of N-004-023 u 6 (20.5g, 68.2mmol) in MeOH (500 mL) in N 2 The mixture was then stirred at 70 ℃ under reflux for 16h and the reaction was treated with water (500 mL). The aqueous phase was extracted with DCM (2 × 300 mL). The combined organic phases were washed with saturated brine (2X 300 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated to purify by silica gel chromatography (PE/EtOAc =10/1 to 6/1) to give N-004-023 u 7 (20g, 88%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.12-5.06(m,1H),3.38(s,3H),3.19(s,2H),2.25-2.22(m,1H),2.20-2.09(m,3H),1.66-1.63(m,3H),1.60-1.24(m,14H),1.22-1.00(m,6H),0.87(s,3H)。
6. At 0 ℃ in N 2 Next, 9-BBN dimer (29.2g, 120mmol) was added to a solution of N-004-023 u 7 (20g, 60.1mmol) in THF (100 mL). The solution was stirred at 65 ℃ for 2 hours. After cooling to 0 ℃ EtOH (34.9mL, 601mmol) was added. NaOH solution (120mL, 5M, 601mmol) was then added very slowly. After addition, H was slowly added 2 O 2 (68.0 g,601mmol,30% in water) and the internal temperature was kept below 10 ℃. The mixture was heated at 75 ℃ under N 2 Stirred for 1 hour. The mixture was cooled to 25 ℃ again. Adding the mixture to H 2 O (2L). The mixture was stirred for 30mins. The precipitate was collected by filtration and washed with H 2 O (2X 500 mL) to give N-004-023 u 8 (17.8g, 85%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.70-3.55(m,1H),3.38(s,3H),3.19(m,2H),2.11-1.86(m,4H),1.80-1.25(m,13H),1.23-0.88(m,12H),0.68(s,3H)。
Figure BDA0003762336660001731
7. Silica gel (24 g) and PCC (24.5 g, 114mmol) were added to a suspension of N-004-023 upu 8 (20g, 57.0mmol) in DCM (500 mL) at 25 ℃. The mixture was stirred at 25 ℃ for 2 hours. The mixture was filtered and the filter cake was washed with DCM (2 × 100 mL). The combined filtrates were concentrated in vacuo. The residue was purified by flash column (0-30% EtOAc in PE) to give NA-004-023_9 (19g, 95%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.39(s,3H),3.20(s,2H),2.60-2.52(m,1H),2.20-2.10(m,5H),1.99-1.80(m,3H),1.75-1.40(m,12H),1.30-1.04(m,7H),0.61(s,3H)。
8. t-BuOK (12.2g, 54.5 mmol) was added to MePPh at 25 deg.C 3 A suspension of Br (38.9 g, 109mmol) in THF (300 mL). After addition, the reaction mixture was heated to 45 ℃ and stirred for 1hr. N-004-023_9 (19g, 35.9mmol) was then added and the reaction mixture was stirred at 45 ℃ for 16 hours. NH for the mixture 4 Cl (100 mL, saturated aqueous solution). The organic layer was separated. The aqueous phase was extracted with EtOAc (2 × 300 mL). The combined organic phases were washed with saturated brine (2X 200 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was taken up in MeOH/H at 25 deg.C 2 O (100 mL/100 mL) was triturated to give N-004-023_10 (17g, 90%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.68(s,1H),3.39(s,3H),3.19(s,2H),2.10-2.04(m,1H),2.03-1.90(m,3H),1.75-1.56(m,12H),1.49-1.25(m,4H),1.22-0.89(m,8H),0.57(s,3H)。
9. 9-BBN dimer (29.5g, 122mmol) was added to a solution of N-004-023 u 10 (17g, 49.0 mmol) in anhydrous THF (300 mL) and at 0 deg.C in N 2 Stirring was continued for 30 minutes. The reaction mixture was warmed to 25 ℃ (room temperature) and stirred for 2 hours. The reaction mixture was cooled. The mixture was quenched by EtOH (100 mL) at 0 ℃. NaOH (98.0mL, 490mol,5M in water) was added very slowly. After addition, H was slowly added 2 O 2 (44.5mL, 490mmol, 11M) until the internal temperature did not rise any more and the temperature remained low during this periodAt 30 ℃. The mixture was stirred at 50 ℃ for a further 1hr. The mixture was then cooled, treated with water (2L) and stirred for 30 minutes. The suspension was filtered under vacuum to give N-004-023_11 (17 g, crude material) as a solid. N-004-023_11 (17g, 46.6 mmol) from MeOH/H 2 O (100/100 mL) was triturated at 25 ℃ and stirred for 1 hour. The suspension was filtered under vacuum to give N-004-023_11 (14 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.79-3.66(m,1H),3.50-3.37(m,4H),3.28(s,2H),2.43(s,1H),2.26-1.98(m,3H),2.18-2.12(m,1H),1.95-1.60(m,11H),1.34-1.04(m,14H),0.76(s,3H)。
10. DMP (9.24g, 21.8mmol) was added to a solution of N-004-023 glu 11 (4g, 10.9mmol) in DCM (80 ml) at 25 ℃. A drop of water was added to the mixture and stirred for 30 minutes. The reaction mixture was saturated NaHCO 3 Aqueous solution pH =7-8 quenched below 10 ℃. The DCM phase in the filtrate was separated and saturated NaHCO was used 3 /Na 2 S 2 O 3 Aqueous solution (1,2x50ml) was washed. The organic phase was washed with saturated brine (2X 50 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-004-023 u 12 (1.8g, 46%) as an oil.
1 H NMR(400MHz,CDCl 3 )δ9.57-9.55(m,1H),3.38(s,3H),3.20(s,2H),2.39-2.26(m,1H),2.17-2.06(m,1H),2.05-1.75(m,4H),1.74-1.53(m,8H),1.85-1.00(m,15H),0.74(s,3H)。
11. CsF (1.86g, 12.3mmol) was added to a solution of N-004-023_12 (1.8g, 4.96mmol) in anhydrous THF (20 mL) at 0 ℃. After stirring at 0 ℃ for 20 minutes, TMSCF was added at 0 DEG C 3 (1.74g, 12.3mmol) and stirring for 1 hour, then TBAF.3H is added 2 O (6.25g, 19.8mmol). The mixture was warmed to 50 ℃ and stirred for an additional 1 hour. The reaction mixture was poured into ice-water (50 mL) and stirred for 10 min. The aqueous phase was extracted with EtOAc (2 × 80 mL). The combined organic phases were washed with saturated brine (2X 80 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-20% EtOAc in PE) to give N-004-023\13 (1.2g, 56%) as an oil.
1 H NMR(400MHz,CDCl 3 )δ4.03-3.98(m,1H),3.39(s,3H),3.20(s,2H),2.17-1.80(m,7H),1.73-1.41(m,10H),1.28-0.95(m,13H),0.71(s,3H)。
12. DMP (2.34g, 5.54mmol) was added to a solution of N-004-023_13 (1.2g, 2.77mmol) in DCM (30 ml) at 25 ℃. After stirring for 30mins at 25 ℃ the reaction mixture was diluted with saturated NaHCO 3 Aqueous solution pH =7-8 (30 mL) quenched below 10 ℃. DCM (30 mL) was then added and the mixture was stirred for 10 min. The suspension was filtered. The DCM phase in the filtrate was separated and saturated NaHCO was used 3 /Na 2 S 2 O 3 Aqueous solution (1, 2x50ml). The organic phase was washed with saturated brine (2 × 50 mL). With anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford N-004-023 u 13a (1.2 g, crude) as an oil.
1 H NMR(400MHz,CDCl 3 )δ3.39(s,3H),3.20(s,2H),3.05-2.95(m,1H),1.91-1.51(m,10H),1.46-1.20(m,10H),1.17-0.96(m,8H),0.72(s,3H)。
13. NaBH is reacted at 0 DEG C 4 (210mg, 5.56mmol) was added to a solution of N-004-023 u 13A (1.2g, 2.78mmol) in MeOH (5 mL) and stirred for 30min. With MeOH/H 2 After O (20/20 mL) treatment, the mixture was stirred for 10 minutes. The aqueous phase was extracted with EtOAc (2 × 50 mL). The combined organic phases were washed with saturated brine (2X 50 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-20% EtOAc in PE) to give 48 (57mg, 5%) and 45 (200 mg, impure) solids. 45 (200mg, 0.462mmol) was purified by flash column (0-30% EtOAc in PE) to give 45 (120mg, 10%) as a solid.
45:
1 H NMR(400MHz,CDCl 3 )δ4.01-3.98(m,1H),3.38(s,3H),3.19(s,2H),2.15-2.10(m,1H),2.05-1.80(m,5H),1.72-1.55(m,5H),1.54-1.34(m,6H),1.31-1.20(m,4H),1.16-0.95(m,9H),0.71(s,3H)。
LCMS Rt =1.129 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, MS ESI C 24 H 38 F 3 [M-HO 3 ] + Calculated value 383 of (d), found value 383.
Example 46: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -17- ((2S, 3R) -4-cyclopentyl-3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (46)
Figure BDA0003762336660001761
1. A solution of (bromomethyl) cyclopentane (2.25g, 13.8mmol) in THF (8 mL) was added dropwise to Mg (662mg, 27.6mmol) and I at 75 deg.C 2 (70mg, 0.276 mmol) in THF (3 mL). The mixture was stirred at 75 ℃ for 1 hour. After cooling, a solution of S-500-6-1_1 (1g, 2.78mmol) in THF (30 mL) was added slowly at 15 ℃. After addition, the mixture was stirred at 15 ℃ for 2hrs with sat 4 Cl (40 mL) and saturated citric acid (20 mL) were quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases were washed with brine (2 × 30 mL) and Na 2 SO 4 Dried, filtered and concentrated and purified by combi-flash (0-15% etoac in PE) to give a mixture of S-500-6-13 \u1 and 22 isomers (900mg, 73%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.32-5.23(m,1H),3.77-3.67(m,1H),2.40-2.30(m,1H),2.07-1.89(m,4H),1.88-1.69(m,4H),1.67-1.59(m,4H),1.55-1.26(m,15H),1.16-1.05(m,5H),1.05-1.00(m,4H),0.99-0.81(m,8H),0.68(s,3H)。
2. DMP (1.72g, 4.06mmol) was added to a solution of S-500-6-13-u 1 (900mg, 2.03mmol) in DCM (30 mL). Then, the reaction mixture was stirred at 15 ℃ for 10 minutes. The reaction mixture was saturated NaHCO 3 The aqueous solution (50 mL) was quenched until the pH of the aqueous layer became about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (20 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3X 40 mL) and saturationNaHCO 3 (40 mL), brine (40 mL), na 2 SO 4 Dried, filtered and concentrated to give crude S-500-6-13-u 2 (900 mg, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.30-5.25(m,1H),2.54-2.42(m,2H),2.41-2.33(m,1H),2.30-2.17(m,1H),2.06-1.90(m,3H),1.87-1.78(m,2H),1.73-1.66(m,2H),1.65-1.35(m,15H),1.33-1.21(m,2H),1.17-0.92(m,13H),0.88-0.82(m,3H),0.69(s,3H)。
3. Reacting NaBH 4 (3.46g, 102mmol) is added five times every 5 minutes to a solution of S-500-6-13-u 2 (900mg, 2.04mmol) in MeOH (5 mL) and THF (5 mL). The mixture was stirred at 15 ℃ for 30 minutes with saturated NH 4 Cl (50 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases are washed with Na 2 SO 4 Drying, filtration, concentration and purification by combi-flash (0-15% EtOAc in PE) to give impure 46 (120 mg) as a solid, which was separated by SFC ((column: AD (250mm 30mm, 5um), gradient: 45-45% B (A =0.05% NH) 3 /H 2 O, B = MeOH), flow rate: 60 mL/min) to give pure 46 (100mg, 84%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.76-3.67(m,1H),2.40-2.33(m,1H),2.08-1.91(m,4H),1.90-1.78(m,2H),1.77-1.55(m,10H),1.54-1.31(m,9H),1.26-1.22(m,2H),1.22-1.05(m,6H),1.03(s,3H),1.01-0.89(m,5H),0.89-0.82(m,3H),0.69(s,3H)。
LCMS Rt =1.474 min, chromatography at 2.0 min, 30-90AB _E, 99% purity, MS ESI C 30 H 49 O[M+H-H 2 O] + Calculated value 425 of (a), measured value 425.
Example 47: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -3,10, 13-trimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (47)
Figure BDA0003762336660001781
1. Pd (OH) 2 (200 mg) was added to a solution of 30 (100mg, 0.239mmol) in MeOH (10 mL). The mixture was heated at 50 ℃ in H 2 Stirring was carried out at 50 Psi. The mixture was filtered, concentrated and purified by combi-flash (0-10% etoac in PE) to give 47 (21mg, 21%) and 36 (1mg, 1%) as white solids.
47:
1 H NMR(400MHz,CDCl 3 )δ3.68-3.54(m,1H),2.02-1.90(m,1H),1.76-1.57(m,6H),1.54-1.27(m,10H),1.26-1.21(m,7H),1.20-1.08(m,5H),1.07-0.95(m,3H),0.94-0.83(m,10H),0.81(s,3H),0.72-0.60(m,4H)。
LCMS t R =1.290 min, chromatography by 2min, 30-90AB _ELSD, purity 100.0%, MS ESI C 28 H 47 [M+H-2H 2 O] + Calculated value 383 of (d), found value 383.
Example 48: synthesis of (3S, 5R,8R,9R,10S,13S,14S, 17R) -3- (methoxymethyl) -13-methyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybutan-2-yl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (48)
Figure BDA0003762336660001791
NaBH is reacted at 0 DEG C 4 (210mg, 5.56mmol) was added to a solution of N-004-023 u 13A (1.2g, 2.78mmol) in 1.MeOH (5 mL) and stirred for 30min. With MeOH/H 2 After O (20/20 mL) treatment, the mixture was stirred for 10 minutes. The aqueous phase was extracted with EtOAc (2 × 50 mL). The combined organic phases were washed with saturated brine (2 × 50 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-20% EtOAc in PE) to give 48 (57mg, 5%) solids and 45 (200 mg, impure) solids. 45 (200mg, 0.462mmol) was purified by flash column (0-30% EtOAc in PE) to give 45 (120mg, 10%) as a solid.
48:
1 H NMR(400MHz,CDCl 3 )δ4.07-4.01(m,1H),3.39(s,3H),3.19(s,2H),2.30-2.22(m,1H),2.14-2.05(m,1H),2.00-1.80(m,4H),1.72-1.57(m,6H),1.49-1.20(m,9H),1.18-0.95(m,9H),0.68(s,3H)。
LCMS Rt =1.085 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, MS ESI C 24 H 38 F 3 [M-HO 3 ] + Calculated value 383 of (d), found value 383.
Example 49: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -17- ((2S, 3S) -4- (4, 4-dimethylcyclohexyl) -3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (49)
Figure BDA0003762336660001801
The synthesis of 49 is described in example 4.
49:
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.85-3.77(m,1H),2.40-2.32(m,1H),2.07-1.87(m,4H),1.76-1.69(m,1H),1.66-1.55(m,5H),1.53-1.42(m,7H),1.41-1.31(m,5H),1.30-1.12(m,8H),1.11-1.05(m,3H),1.03(s,3H),1.01-0.92(m,2H),0.91-0.82(m,12H),0.68(s,3H)。
LCMS Rt =1.718 min, chromatography at 2.0 min, 30-90AB _E, 98% purity, MS ESI C 33 H 53 [M+H-2H 2 O] + Calculated 449, found 449.
Example 50: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxybut-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (50)
Figure BDA0003762336660001802
Figure BDA0003762336660001811
1. At 0 ℃ in N 2 MeMgBr (0.83mL, 2.49mmol,3M in ether) was added dropwise to a solution of N-8-7/u 1 (300mg, 0.832mmol) in THF (20 mL). After stirring for 30 minutes at 20 ℃, the reaction was quenched with saturated NH 4 Cl (50 mL) quenched and extracted with EtOAc (2 × 10 mL). The combined phases were washed with brine (10 mL) and Na 2 SO 4 Dried, filtered, concentrated and purified by flash column (step 2) (0-10% EtOAc in PE) to give 50 (40mg, 29%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.98-3.89(m,1H),1.99-1.84(m,2H),1.69-1.56(m,6H),1.54-1.45(m,2H),1.43-1.29(m,6H),1.28-1.17(m,4H),1.17-1.12(m,4H),1.12-0.94(m,5H),0.92-0.84(m,7H),0.82(s,3H),0.68-0.61(m,4H)。
LCMS Rt =3.428 min, chromatography at 7.0 min, 30-90AB _E, 100% purity, MS ESI C 25 H 41 [M+H-2H 2 O] + The calculated value 341, found value 341.
Example 51: synthesis of 3S,5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -4- (4, 4-dimethylcyclohexyl) -3-hydroxybut-2-yl) -3- (methoxymethyl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (51)
Figure BDA0003762336660001821
1. At 25 ℃ in N 2 Next, S-500-6-29-u 2 (999mg, 1.22M in THF, 4.87 mmol) was added dropwise to a solution of N-8-1-u 1 (210mg, 05576mmol) in THF (2 mL). After stirring at 25 ℃ for 16hrs, the reaction mixture was saturated with NH 4 Cl (10 mL) quenched and extracted with ethyl acetate (3 × 10 mL). The combined organic layers were washed with brine (30 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product, which was purified by flash column (0-15% etoac in PE) 2 times to give an impure product (30 mg). The impure product was further purified by ELSD prep-HPLC (column: phenomenex Synergi C18 x 30mm x 4um), gradient: 90-95% B (A = water (0.05% HCl)), B =MeCN), flow rate: 25 mL/min) to give pure 51 (4 mg,1.4% yield) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.84-3.76(m,1H),3.45-3.32(m,5H),2.62-2.39(m,1H),1.99-1.85(m,2H),1.73-1.62(m,4H),1.53-1.40(m,7H),1.39-1.31(m,5H),1.30-1.21(m,7H),1.20-1.13(m,4H),1.12-1.10(m,5H),0.99-0.93(m,1H),0.89-0.86(m,6H),0.85(s,3H),0.83(s,3H),0.68-0.61(m,4H)。
LCMS Rt =5.669 min, chromatography over 7.0 min, 30-90AB _E, 100% purity, MS ESI C 33 H 55 O[M+H-2H 2 O] + Found 467 in the calculation of (1).
Example 52: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-6-methylheptan-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (52)
Figure BDA0003762336660001831
1. NaOH solution (974 mg in 6mL H 2 O, 16.8 mmol) was added to a solution of S-500-2-15_1 (900mg, 1.68mmol) in THF (10 mL) and MeOH (5 mL). The mixture was heated at 50 ℃ for 16 hours. Saturated NH for reaction mixture 4 Cl (60 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases were washed with brine (60 mL) and Na 2 SO 4 Dried, filtered, concentrated, and purified by combi-flash (0-15% etoac in PE) to give 210mg solid, which was purified by SFC (column: AD (250mm × 30mm,5 um), gradient: 35-35% b (a =0.1% nh% 3 /H 2 O, B = MeOH), flow rate: 80mL/min) to give 52 (150mg, 68%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.30-5.26(m,1H),3.64-3.58(m,1H),2.40-2.30(m,1H),2.02-1.92(m,3H),1.80-1.58(m,7H),1.56-1.31(m,9H),1.30-1.05(m,8H),1.03(s,3H),1.02-0.96(m,2H),0.95-0.86(m,9H),0.85-0.80(m,3H),0.69(s,3H)。
LCMS t R =1.335 min, chromatography over 2 min, 30-90ab _elsd, purity 100.0%, MS ESI C 29 H 47 [M+H-2H 2 O] + Calculated 395, found 395.
Example 53: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -10, 13-dimethyl-17- ((2S, 3R) -4, 4-trifluoro-3-hydroxybut-2-yl) -3- (trifluoromethyl) -2,3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (53)
Figure BDA0003762336660001841
1. To a solution of N-004-027_1 (1.5g, 3.76mmol) in anhydrous THF (40 mL) at 0 deg.C was added CsF (1.42g, 9.40mmol). After stirring at 0 ℃ for 20 minutes, TMSCF was added at 0 DEG C 3 (1.33g, 9.40mmol) and stirred for 30 minutes. The color changed to light yellow. Addition of TBAF.3H 2 O (4.74g, 15.0 mmol) and stirred at 50 ℃ for 30 minutes. The reaction mixture was poured into ice-water (100 mL). The aqueous phase was extracted with EtOAc (2 × 100 mL). The combined organic phases were washed with saturated brine (2X 100 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated to give the isomer mixture (1.45 g, crude) as a yellow solid which was purified by flash column (0-15% etoac in PE) to give white solid 53 (340mg, 24%) and white solid 1 (200mg, 14%).
53:
1 H NMR(400MHz,CDCl 3 )δ5.38-5.36(m,1H),4.06-3.94(m,1H),2.49(s,2H),2.09-1.58(m,13H),1.48-0.85(m,14H),0.73(s,3H)。
LCMS Rt =1.134 min, chromatography over 2min, 30-90AB _2MIN _E, 99% pure,
MS 50-100_1_4min. M for C 24 H 33 F 6 O[M+H-H 2 O] + 451, found 451.
Example 54: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-6, 6-dimethylhept-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (54)
Figure BDA0003762336660001851
1. NaBH is added at 15 ℃ 4 (1.77g, 46.8mmol) was added portionwise to a solution of S-500-6-1_3 (520mg, 1.17mmol) in THF (5 mL) and MeOH (10 mL). The mixture was stirred at 15 ℃ for 20 minutes. The mixture is treated with NH 4 Cl (20 mL, saturated aqueous) was quenched and extracted with EtOAc (50 mL). The organic layer was separated and concentrated in vacuo to give a mixture which was subjected to flash column separation (0-15% etoac in PE) to give S-500-6-1 (300 mg, impure) and 54 (170 mg, impure).
Impure 54 (220 mg, impure) was purified by flash column (0-15% etoac in PE) to give a solid. The solid was dissolved in MeCN (50 mL) at 60 ℃ and concentrated in vacuo to give 54 (120mg, 23%) as a solid.
54:
1 H NMR(400MHz,CDCl 3 )δ5.33-5.24(m,1H),3.62-3.52(m,1H),2.42-2.31(m,1H),2.11-1.90(m,3H),1.72-1.35(m,15H),1.29-1.08(m,8H),1.03(s,3H),1.01-0.96(m,2H),0.93(d,J=6.8Hz,3H),0.90(s,9H),0.85(t,J=7.6Hz,3H),0.70(s,3H)。
LCMS Rt =5.463 min, chromatography at 7.0 min, 30-90_AB _E, 100% purity, MS ESI C 30 H 49 [M+H-2H 2 O] + The calculated value 409 of (1) is actually measured value 409.
Example 55: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((1R, 2S) -1-hydroxy-1-phenylprop-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (55)
Figure BDA0003762336660001861
1. A solution of N-8-7_1 (300mg, 0.832mmol) in THF (5 mL) was added to PhMgBr (1.38mL, 3M in ether at 0 deg.C,4.15 mmol) in THF (10 mL) and the reaction mixture was then stirred at 0 ℃ for 3 hours. Then, the reaction mixture was stirred at 25 ℃ for 5 hours. The reaction mixture was quenched by water (10 mL) at 0 ℃. The solution was filtered and the filter cake was washed with EtOAc (10 mL). The aqueous phase was extracted with EtOAc (3X 15 mL). The combined organic phases were washed with saturated brine (2X 10 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE/EtOAc =20/1 to 1/1) to give 59 and 19 (200 mg, crude material) as solids. The crude product was purified by SFC (column: AD (250mm 30mm, 5um)), gradient: 25-25% B (a =0.1% nh3/H2O, B = EtOH), flow rate: 60 mL/min) to give 55 (Peak 2, 55mg, 15%) and 19 (Peak 1, 21mg, 6%) as solids.
55:
1 H NMR(400MHz,CDCl 3 )δ7.38-7.28(m,4H),7.25-7.20(m,1H),δ4.95-4.90(m,1H),2.13-2.01(m,1H),1.98-1.88(m,1H),1.77-1.59(m,6H),1.57-1.43(m,6H),1.43-0.93(m,13H),0.91-0.85(m,3H),0.83(s,3H),0.76-0.72(m,3H),0.68(s,4H)。
LCMS Rt =1.239 min, chromatography at 2.0 min, 30-90ab \u2 min. Purity 100%, MS ESI C 30 H 43 Calculated value of [ M-2H ] 2 O+H] + 403, measured value 403.
SFC Rt =1.192 min, with 3 min chromatography, OJ _ 3. Mu. EtOH _. Mu. DEA _. Mu. 5. Mu. 40. Mu. 25ML,99% de.
Example 56: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3-Ethyl-10, 13-dimethyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybutan-2-yl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (56)
Figure BDA0003762336660001871
1. To a solution of 81 (1 g, 3.26mmol) in MeOH (30 mL) and THF (10 mL) was added Pd (OH) 2 (1g,<1% water). The mixture was then hydrogenated at 50psi at 50 ℃ for 48 hours. The mixture was filtered through a pad of celite without monitoring and the filtrate was concentrated in vacuo. Residue ofPurification by flash column (PE/EtOAc =10/1 to 5/1) gave 56 (331mg, 33%) as a white solid.
1 H NMR(400MHz,CDCl3)δ4.09-3.99(m,1H),2.18-2.13(m,1H),1.99-1.78(m,4H),1.75-1.59(m,3H),1.50-1.3(m,7H),1.34-1.22(m,6H),1.21-1.00(m,10H),0.96(s,3H),0.94-0.89(m,3H),0.67(s,3H)。
LCMS Rt =1.184 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 25 H 40 F 3 O[M+H-H 2 O] + Calculated value 413, found value 413.
Example 57: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3,10, 13-trimethyl-17- ((2S, 3R) -4, 4-trifluoro-3-hydroxybutan-2-yl) hexahydro-1H-cyclopenta [ a ] phenanthren-3-ol (57)
Figure BDA0003762336660001881
1. In N 2 A suspension of LiCl (13.9g, 329mmol, anhydrous) in THF (500 mL, anhydrous) was stirred at 10 ℃ for 30mins. FeCl addition at 10 deg.C 3 (27.8 g,172mmol, anhydrous). The mixture was cooled to-30 ℃. MeMgBr (209mL, 3M in ether) was added dropwise to the mixture at-30 ℃. After stirring at-30 ℃ for 10mins, S-500-2-12 (50g, 157mmol) was added at-30 ℃. The mixture was stirred at-15 ℃ for 2 hours and quenched with citric acid (500ml, 10% aq.). The mixture was extracted with EtOAc (3 × 800 mL). The combined organic phases were washed with saturated brine (300 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product, which was purified by silica gel column (PE/DCM/EtOAc = 1/1/1) to give S-500-2-12 \u2 (50g, 86%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.57-2.48(m,1H),2.23-2.13(m,1H),2.06-1.78(m,3H),1.64-1.25(m,14H),1.24-1.01(m,10H),0.96(s,3H),0.74(s,1H),0.60(s,3H)。
2. To PPh at 20 ℃ 3 To a suspension of MeBr (79.7g, 244mmol) in THF (400 mL) was addedt-BuOK (25.1g, 224mmol). After stirring for 30 min at 40 ℃, a solution of S-500-2-12_2 (50g, 150mmol) in THF (100 mL) was added at 40 ℃ and the reaction mixture was stirred for 1 h at 40 ℃. The reaction mixture was poured into 50g of ice and stirred for 15 minutes. The organic layer was separated and the aqueous phase was extracted with EtOAc (3 × 50 mL). The combined organic phases were concentrated in vacuo to give an oil. Crude product in MeOH/H 2 Trituration in O (200 mL/200 mL) and filtration to give S-500-2-12 \u3 (55g, 88%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.69(s,1H),2.06-1.79(m,4H),1.75(s,3H),1.73-1.58(m,4H),1.56-1.25(m,9H),1.22(s,3H),1.21-1.02(m,6H),1.01-0.94(s,3H),0.93-0.74(m,1H),0.55(s,3H)。
3. At 25 ℃ in N 2 9-BBN dimer (60.7g, 249mmol) was added and stirred for 1 hour to a solution of S-500-2-12_3 (55g, 166mmol) in THF (500 mL) to form a solid. Ethanol (95.3 mL, 1.66mol) and NaOH (166mL, 5M, 830mmol) were added to the reaction mixture. The mixture became clear. H is added dropwise at 25 DEG C 2 O 2 (132mL, 10M, 1.32mol) and the internal temperature was raised to reflux (75 ℃). After addition the mixture was cooled and stirred for 16hrs to form a solid. Na was added to the mixture at 25 ℃ 2 S 2 O 3 (500mL, 20% aq.) and water (1L). The mixture was stirred for 1 hour. Upon turning off the agitator, clear lower and upper suspension layers were formed. The clear lower layer was discarded. Water (2L) was added to the upper suspension layer. The mixture was stirred for 15mins. The mixture was filtered to give S-500-2-12 \u4 (50 g, impure) as a solid. S-500-2-12-u 4 (50g, 143mmol, impure) was placed in EtOH/H 2 Trituration in O (90 mL/10 mL) at 100 ℃ for 2 hours, then cooling to 15 ℃ and filtration to give S-500-2-12 \u4 (38 g, impure) as a solid. S-500-2-12 (38g, 109mmol, impure) was placed in EtOH/H 2 Trituration in O (45 mL/5 mL) at 100 ℃ for 2 hours, then cooling to 15 ℃ and filtration to give S-500-2-12 \u4 (28g, 43%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.67-3.59(m,1H),3.39-3.32(m,1H),2.01-1.75(m,4H),1.69-1.58(m,3H),1.54-1.24(m,10H),1.23-1.14(m,9H),1.09-1.02(m,5H),0.96(s,3H),0.74(m,1H),0.67(s,3H)。
4. To a solution of N-004-016-u 1 (10.0 g,28.6 mmol) in DCM (100 mL) was added DMP (24.2g, 57.2mmol). Then H is mixed 2 O (0.2 mL) was added to the mixture. Then, the reaction was stirred at 25 ℃ for 1 hour. Adding saturated NaHCO to the reaction mixture 3 Aqueous solution (100 mL). The mixture was filtered and the filter cake was washed with DCM (2 × 100 mL). The mixture was liquid and separated, and the aqueous phase was extracted with DCM (2 × 100 mL). The combined organic layers were washed with saturated NaHCO 3 Aqueous solution/Na 2 S 2 O 3 (100 mL/100 mL) and brine (100 mL), washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford a white solid. The residue was purified by silica gel chromatography (PE/EtOAc =0 to 20%) to give N-004-016_2 (3.5g, 35%) as a white solid.
1 H NMR(400MHz,CDCl3)δ9.58-9.54(m,1H),2.39-2.32(m,1H),1.96-1.77(m,4H),1.69-1.31(m,14H),1.23-1.16(m,6H),1.14-1.02(m,5H),0.96(s,3H),0.76-0.59(m,4H)。
5. To a solution of N-004-016 (1.5g, 4.32mmol), csF (328mg, 2.16mmol) in THF (10 mL) at 0 deg.C was added TMSCF 3 (1.53g, 10.8mmol). The mixture was stirred at 25 ℃ for 1 hour. TBAF was added to the mixture. 3 H 2 O (3.4g, 10.8mmol). The mixture was stirred at 25 ℃ for 2 hours. The mixture was quenched with water (20 mL) and extracted with EtOAc (2 × 30 mL). The combined organic phases were washed with brine (50 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by flash column (0-15% EtOAc in PE) to give N-004-017_1 (700mg, 39%) as a white solid.
1 H NMR(400MHz,CDCl3)δ4.07-3.96(m,1H),2.21-2.09(m,1H),1.99-1.77(m,5H),1.72-1.29(m,15H),1.24-1.20(m,4H),1.13-1.01(m,5H),0.96(s,3H),0.89-0.84(m,1H),0.76-0.64(m,3H),0.60(s,1H)。
6. To N-004-017 (700mg, 1.68mmo) 1 at 25 deg.Cl) to a solution in pyridine (5 mL) were added benzoyl chloride (354 mg, 2.52mmol) and DMAP (102mg, 0.84mmol). The mixture was heated to 60 ℃ and stirred for 10 hours. The reaction mixture was diluted with EtOAc (10 mL) and then quenched with water (10 mL). The aqueous was extracted with EtOAc (2 × 20 mL). The combined organic layers were washed with brine (20 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford a white solid. The residue was purified by flash column (0-15% EtOAc in PE) to give N-004-017_2 (600mg, 68%) as a white solid.
LCMS Rt =1.464 min, chromatography over 2min, 30-90ab _2min _e.m, purity 92%.
SFC conditions Peak 1: rt =2.434 minutes and peak 2: rt =2.541 min, 8min chromatography, OD _ MEOH (DEA) _5_40_2,8ML _8MIN.M (column: chiralcel OD-3X 4.6mm I.D.,3um mobile phase: A: CO 2B: methanol (0.05% DEA) gradient: 5% to 40% B was maintained within 4.5min and at 40% for 2.5 min, then 5% B was maintained for 1 min, flow rate: 2.8mL/min, column temperature: 40 ℃).
7.N-004-017 u 2 (600mg, 1.15mmol) purified by SFC (column: chiralcel OD (250mm 30mm, 5um), gradient: 20-20% B (A =0.1% NH) 3 /H 2 O, B = MeOH), flow rate: 60 mL/min) to yield N-004-017_3 (peak 2, 190mg, impure, 31%), N-004-018 \u1 (peak 1, 180mg, 30%) as a white solid. Impure N-004-017 [ u 3 ] (190mg, 0.36mmol) was purified by SFC (column: OD (250mm 30mm, 5um), gradient: 20-20% by B (A =0.1% 3 /H 2 O, B = MeOH), flow rate: 60 mL/min) to give N-004-017_3 (100mg, 53%) as a white solid.
N-004-018_1:
1 H NMR(400MHz,CDCl3)δ8.12-8.06(m,2H),7.65-7.59(m,1H),7.53-7.46(m,2H),5.68-5.58(m,1H),2.15-2.03(m,2H),1.97-1.69(m,3H),1.67-1.57(m,3H),1.44-1.24(m,9H),1.23-1.13(m,11H),1.12-1.97(m,3H),0.94(s,3H),0.72(s,3H)。
LCMS Rt =1.525 min, chromatography over 2min, 30-90ab _2min _ &220 &254.Lcm, 100% purity.
SFC _ D1 Rt =2.450 min, OD _ MEOH (DEA) _5_40_2,8ML _8MIN.M,100% de by 8min chromatography.
N-004-017_3:
1 H NMR(400MHz,CDCl3)δ8.14-8.06(m,2H),7.64-7.57(m,1H),7.52-7.44(m,2H),5.63-5.52(m,1H),2.11(s,1H),2.06-1.77(m,6H),1.72-1.62(m,3H),1.44-1.32(m,8H),1.28-1.18(m,12H),0.99-0.93(m,4H),0.65(s,3H)。
LCMS Rt =1.529 min, chromatography over 2min, 30-90ab _2min _ &220 &254.Lcm, 100% purity.
SFC Rt =2.544 min, OD _ MEOH (DEA) _5_40_2,8ML _8MIN.M,98% de by 8min chromatography.
8. To a solution of N-004-018 \ u 1 (180mg, 0.34mmol) in THF (3 mL) and MeOH (1.5 mL) and water (1.5 mL) was added KOH (96.5mg, 1.72mmol). The mixture was stirred at 60 ℃ for 16 hours. The mixture was poured into water (20 mL) and extracted with EtOAc (2 × 40 mL). The combined organic layers were washed with brine (30 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-15% EtOAc in PE) to give 57 (114mg, 79%) as a white solid.
1 H NMR(400MHz,CDCl3)δ4.00(brs,1H),2.03-1.77(m,8H),1.69-1.61(m,2H),1.54-1.49(m,1H),1.47-1.24(m,10H),1.22(s,3H),1.21-1.10(m,4H),1.08-1.01(m,4H),0.96(s,3H),0.69(s,3H)。
LCMS Rt =1.169 min, chromatography over 2min, 30-90AB (u 2MIN) E.M, 95% purity, MS ESI C 24 H 38 F 3 O[M+H-H 2 O] + Calculated value 399, found value 399.
HPLC Rt =5.44 min, 100% purity at 10 min Ultimate C18 × 50mm 3um, 30-90\uab _1.2ml _e.met.
Example 58: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((1R, 2S) -1-cyclopentyl-1-hydroxypropan-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (58)
Figure BDA0003762336660001921
Figure BDA0003762336660001931
1. At 0 ℃ in N 2 Next, a solution of N-8-7 (500mg, 1.38mmol) in THF (5 mL) was added to cyclopentyl magnesium bromide (1.38mL, 3M in THF). After stirring at 15 ℃ for 18hrs, the reaction mixture was saturated with NH 4 Cl (10 mL) quenched and extracted with EtOAc (2 × 10 mL). The combined organic layers were washed with brine (10 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-10% EtOAc in PE) to give N-8-15 \u1 (170mg, 29%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.39-3.22(m,1H),2.00-1.81(m,4H),1.70-1.41(m,12H),1.41-1.13(m,13H),1.13-0.95(m,6H),0.95-0.79(m,11H),0.65(s,3H)。
2. DMP (0.881g, 2.08mmol) was added to a solution of N-8-15_1 (300mg, 0.696mmol) in DCM (20 mL). After stirring for 10 min at 15 ℃, the reaction mixture was washed with sat 3 (10 mL) quench. The mixture was extracted with DCM (3 × 20 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Washed (3X 20 mL) and brine (20 mL) with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-20% etoac in PE) to give N-8-15_2 (240 mg) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.98-2.89(m,1H),2.62-2.55(m,1H),1.98-1.87(m,1H),1.81-1.72(m,4H),1.71-1.49(m,10H),1.41-1.29(m,4H),1.29-1.19(m,6H),1.14-0.98(m,9H),0.94-0.87(m,4H),0.82(s,3H),0.67(m,5H)。
3. Reacting NaBH 4 (550mg, 14.5 mmol) was added to a mixture of N-8-15_2 (240mg, 0.559mmol) in MeOH (3 mL) and THF (2 mL). The mixture was stirred at 15 ℃ for 0.5h. Add another batch of NaBH 4 (550mg, 14.5mmol). The reaction mixture is stirred againAnd (5) 1h. Water (5 mL) was added to the reaction mixture. The resulting mixture was extracted with EtOAc (2 × 10 mL). The combined organic layers were washed with brine (10 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-5% etoac in PE) to give 58 (7 mg, 5%) as a solid, and 78 (50 mg, impure) was further purified by flash column (0-5% etoac in PE) to give 78 (17mg, 12%) as a solid.
58:
1 H NMR(400MHz,CDCl 3 )δ3.64-3.59(m,1H),2.09-1.90(m,2H),1.89-1.70(m,4H),1.70-1.45(m,11H),1.45-1.32(m,5H),1.32-1.19(m,9H),1.19-1.08(m,3H),1.08-0.98(m,5H),0.98-0.89(m,4H),0.84(s,3H),0.68(s,3H)。
LCMS Rt =4.832 min, chromatography over 7min, 30-90AB 7MIN \ u E, 100% purity, MS ESI C 29 H 47 [M+H-2H 2 O] + 395, found 395.
HPLC Rt =6.338 min, chromatography over 10min, 50-100ab 10min.m, 98% purity.
Example 59: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -4- (4, 4-dimethylcyclohexyl) -3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (59)
Figure BDA0003762336660001951
1. Pd (OH) 2 (200 mg, anhydrous) was added to a solution of S-500-6-30 (140mg, 0.288mmol) in MeOH (30 mL). The mixture was heated at 50 ℃ in H 2 Next (50 Psi) was stirred for 48 hours. The mixture was filtered, concentrated and purified by combi-flash (0-15% EtOAc in PE) to give S-500-6-25 (27mg, 19%) and S-500-6-26 (42mg, 30%) as solids.
S-500-6-25:
1 H NMR(400MHz,CDCl 3 )δ3.84-3.77(m,1H),1.99-1.84(m,2H),1.81-1.72(m,1H),1.68-1.56(m,4H),1.53-1.43(m,5H),1.42-1.32(m,9H),1.31-1.23(m,5H),1.22-.12(m,7H),1.12-1.00(m,5H),0.99-0.95(m,4H),0.94-0.85(m,12H),0.66(s,3H)。
LCMS Rt =1.797 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 33 H 55 [M+H-2H 2 O] + Calculated value of (451), found value of 451.
Example 60: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-ethyl-17- ((2S, 3R) -3-hydroxy-4- ((1R, 2S) -2-methylcyclopropyl) butan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (60)
Figure BDA0003762336660001961
1. To a solution of N-8-7_2 (1g, 2.41mmol) in THF (50 mL) at 0 deg.C was added LiAlH 4 (914mg, 24.1mmol). The grey suspension was heated at 66 ℃ for 18 hours. The reaction mixture was cooled to 0 ℃ and quenched successively by ice-water (914 mg), 15% w/w aqueous NaOH solution (914 mg) and water (2.74 g). The mixture was filtered and the filter cake was washed with DCM (3 × 50 mL). The filtrate was concentrated to give a residue which was purified twice by flash chromatography (ethyl acetate 10% in PE) to give N-8-7_3 (192mg, 19%) and N-8-7_3a (3976 mg, 39%) as an oil.
N-8-7_3:
1 H NMR(400MHz,CDCl3)δ5.59-5.36(m,2H),3.69-3.61(m,1H),2.25-2.12(m,1H),2.08-1.81(m,3H),1.68(d,J=10.0Hz,3H),1.64-1.54(m,9H),1.53-1.15(m,11H),1.14-0.92(m,5H),0.92-0.85(m,5H),0.83(s,4H),0.69-0.60(m,4H)。
N-8-7_3A:
1 H NMR(400MHz,CDCl 3 )δ5.62-5.37(m,2H),3.62(br d,J=10.0Hz,1H),2.20-2.06(m,1H),1.99-1.61(m,6H),1.61-1.44(m,11H),1.43-1.18(m,5H),1.16-0.94(m,6H),0.94-0.85(m,5H),0.82(s,5H),0.70-0.58(m,6H)。
2. Diethyl Zinc (1M in 0 ℃ C.) over 15 minutesToluene, 4.31mL, 4.31mmol) to a solution of DCM (15 mL) at 0 deg.C CH was added 2 I 2 (2.31g, 8.63mmol). The milky suspension was stirred at 0 ℃ for 10 minutes and a solution of preformed Charette ligand ((4R, 5R) -2- (tert-butyl) -N4, N4, N5, N5-tetramethyl-1, 3, 2-dioxaborolan-4, 5-dicarboxamide) (233mg, 0.8638mmol) and N-8-7 \ u 3A (300mg, 0.7199mmol) in DCM (20 ml) was added rapidly by syringe whereupon the reaction mixture became clear. The solution was brought to 25 ℃ and stirred at this temperature for 16h. The reaction was then quenched by addition of saturated NH 4 Aqueous Cl (150 ml) was quenched, the phases separated and the aqueous phase extracted with DCM (3 × 100 ml). The combined organic phases were washed with saturated NaHCO 3 Aqueous solution (150 mL), saturated Na 2 S 2 O 3 Washed with aqueous solution (150 mL), brine (100 mL), na 2 SO 4 Dried, filtered and concentrated under reduced pressure and the residue purified by flash column chromatography (11% ethyl acetate in PE) to give N-8-7_4a (140mg, 45%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.81-3.72(m,1H),1.97-1.90(m,1H),1.76-1.58(m,4H),1.53-1.44(m,4H),1.43-1.15(m,10H),1.13-0.91(m,8H),0.90-0.78(m,12H),0.66(s,3H),0.54-0.34(m,4H),0.32-0.22(m,2H),0.19-0.12(m,1H)。
3. To a solution of N-8-7_4A (140mg, 0.325mmol) in pyridine (5 mL) at 25 ℃ was added benzoyl chloride (91.3mg, 0.65mmol) followed by DMAP (15.8mg, 0.13mmol). The reaction mixture was stirred at 60 ℃ for 16 hours. The reaction mixture was diluted with DCM (80 mL). DCM phase was washed with water (100 mL), 1.0M aqueous HCl (2X 100 mL), 10% NaHCO 3 Aqueous (2X 100 mL), brine (100 mL), washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give an oil which was purified by flash column (1% ethyl acetate in PE) to give N-8-7-u 5a (180 mg, impure) as an oil which was further purified by flash column (PE) to give N-8-7-u 5a (110mg, 61%) as a solid.
LCMS Rt =1.439 min, chromatography over 2 min, 5-95AB \u220&254, purity 93%, MS ESI C 36 H 53 O 2 [M-H 2 O+H] + Calculated value of 517.8, found value of 517.8.
SFC peak 1: rt =4.079 min and peak 2: rt =4.345 min, AD _3 \ u etoh _dea5 _40_25ml ("Chiralpak AD-3 × 4.6mm i.d.,3um mobile phase: a: CO 2B: ethanol (0.05% dea) gradient: 5% to 40% B in 5 min and held at 40% for 2.5 min, then 5% B held for 2.5 min, flow rate: 2.5mL/min, column temperature: 35 ℃".) by 10 min chromatography.
N-8-7A _5 (110mg, 0.206mmol) was purified by SFC (column: AD (250mm. 30mm,5 um)), gradient: 30-30% B (A =0.1% NH) 3 /H 2 O, B = EtOH), flow rate: 60 mL/min) to yield impure N-8-7a _6 (peak 1, 54mg, 50%) as a solid and impure N-8-8a _1 (peak 2, 23mg, impure) as a solid.
SFC Rt =4.088 min, AD _ 3. Mu. EtOH _DEA5. Mu. 40. Mu. 25ML in 10 min chromatography, 100% w/w.
5. To a solution of N-8-7A _6 (54mg, 0.101mmol) in THF/MeOH (1.5 mL/1.5 mL) was added a solution of KOH (45.2mg, 0.807mmol) in water (0.5 mL). The reaction mixture was stirred at 50 ℃ for 16 hours. To this mixture was added HCl (0.2M, 50mL). The suspension was extracted with DCM (2 × 60 mL). The combined organic phases were then treated with 3% NaHCO 3 Aqueous solution (80 mL), brine (80 mL), washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solid which was purified by flash chromatography (15% ethyl acetate in PE) to give 60 (21mg, 48%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.80-3.75(m,1H),1.97-1.91(m,1H),1.76-1.58(m,7H),1.54-1.27(m,7H),1.26-1.06(m,7H),1.04(d,J=6.0Hz,4H),0.95(s,3H),0.90-0.84(m,8H),0.82(s,4H),0.66(s,3H),0.64-0.59(m,1H),0.52-0.37(m,2H),0.32-0.22(m,2H)。
LCMS Rt =1.327 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, MS ESI C 29 H 49 O[M-H 2 O+H] + Calculated value of 413.4, found value of 413.4.
Example 61: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-Ethyl-10, 13-dimethyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybut-2-yl) -2,3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (61)
Figure BDA0003762336660001991
1. DMP (1.22g, 2.88mmol) was added to a solution of S-500-6-11_1 (580 mg, 1.44mmol) in DCM (30 mL). Then, the reaction mixture was stirred at 15 ℃ for 10 minutes. The reaction mixture was saturated with NaHCO 3 The aqueous solution (50 mL) was quenched until the pH of the aqueous layer became about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (20 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3 × 40 mL), sat 3 (40 mL), brine (40 mL), washed with Na 2 SO 4 Dried, filtered and concentrated to give crude S-500-6-11_1a (550 mg, crude material) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ5.34-5.25(m,1H),2.58-2.29(m,4H),2.08-1.90(m,3H),1.78-1.56(m,9H),1.54-1.35(m,6H),1.31-1.21(m,2H),1.19-1.08(m,5H),1.06-0.99(m,5H),0.93-0.82(m,6H),0.69(s,3H)。
2. Reacting NaBH 4 (1.39g, 41.1mmol) was added to a solution of S-500-6-11A (550mg, 1.37mmol) in THF (4 mL) and MeOH (2 mL) at five 2-minute intervals. The mixture was stirred at 15 ℃ for 30 minutes. Mixing the mixture with sat 4 Cl (20 mL) quenched and extracted with EtOAc (3 × 6 mL). The combined organic phases are washed with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give impure 61 (120 mg) as a white solid, which was again further purified by combi-flash (0-15% etoac in PE) to give pure 61 (150mg, 75%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ5.31-5.25(m,1H),3.73-3.59(m,1H),2.44-2.29(m,1H),2.08-1.92(m,3H),1.76-1.57(m,6H),1.54-1.26(m,10H),1.25-1.18(m,3H),1.17-1.06(m,4H),1.03(s,3H),1.00-0.88(m,8H),0.87-0.82(m,3H),0.69(s,3H)。
LCMS Rt =1.345 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 27 H 45 O[M+H-H 2 O] + The calculated value 385 of (1), the measured value 385.
Example 62: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-ethyl-10, 13-dimethyl-17- ((2S, 3R) -4, 4-trifluoro-3-hydroxybut-2-yl) -2,3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (62)
Figure BDA0003762336660002001
1. To a solution of S-500-6-1_1 (500mg, 1.39mmol) and CsF (105mg, 695umol) in THF (5 mL) at 0 deg.C was added TMSCF 3 (493mg, 3.47mmol). The mixture was stirred at 25 ℃ for 1hr and washed with TBAF.3H 2 O (1.09g, 3.47mmol). The mixture was stirred at 25 ℃ for 2hrs, quenched with water (100 mL) and extracted with EtOAc (2 × 50 mL). The combined organic phases were washed with brine (100 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, PE/EA = 10/1) to give S-500-6-1 \u2 (400mg, 67%) as a white solid.
1 H NMR(400MHz,CDCl3)δ5.33-5.24(m,1H),4.06-4.00(m,1H),2.38-2.35(m,1H),2.08-1.82(m,6H),1.77-1.69(m,1H),1.62-1.20(m,13H),1.16-1.00(m,8H),0.99-0.92(m,1H),0.87-0.83(m,4H),0.74-0.64(m,3H)。
2.3.5 g S-500-6-1 \, 2 by SFC separation (column: AD (250mm 30mm,5 um), gradient: 40-40% B (A =0.05% 3 /H 2 O, B = MeOH), flow rate: 200 mL/min) to give pure 81 (1g, 28%, peak 1) and 62 (1871mg, 53%, peak 2) as white solids.
62:
1 H NMR(400MHz,CDCl3)δ5.30-5.28(m,1H),4.03-3.99(m,1H),2.38-2.34(m,1H),2.10-1.83(m,6H),1.78-1.55(m,5H),1.52-1.32(m,6H),1.31-1.01(m,12H),0.98-0.92(s,1H),0.85(t,J=8Hz,3H),0.73(s,3H)。
LCMS Rt =1.219 min, 2.0 min chromatography, 30-90AB, 100% purity, MS ESI C 25 H 38 F 3 Calculated value of O [ M + H-H 2 O] + 411, found 411.
SFC peak 2: <xnotran> Rt =5.262 , 10 , AD _3_EtOH_DEA_5_40_25ML,99%de. </xnotran>
Example 63: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-ethyl-17- ((2S, 3S) -3-hydroxy-4- (tetrahydro-2H-pyran-4-yl) but-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (63)
Figure BDA0003762336660002021
1. Water (1 mL) and KOH (177mg, 3.17mmol) were added to a solution of N-6-10_1 (180mg, 0.318mmol) in THF (2 mL) and methanol (1 mL). The mixture was stirred at 50 ℃ for 18 hours. The reaction mixture was cooled, diluted with water (5 mL), acidified with 10% hcl (0.2 mL) and extracted with EtOAc (3 × 5 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by column chromatography on silica gel (10-30% etoac in PE) to give 63 (108mg, 74%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.98-3.90(m,2H),3.85-3.77(m,1H),3.44-3.33(m,2H),1.99-1.82(m,2H),1.69-1.58(m,6H),1.57-1.45(m,6H),1.43-1.29(m,7H),1.28-1.14(m,7H),1.13-0.95(m,5H),0.93-0.84(m,7H),0.83(s,3H),0.69-0.61(m,4H)。
LCMS Rt =1.167 min, chromatography at 2.0 min, 30-90AB, purity 100%.
MS ESI C 30 H 49 O[M-2H 2 O+H] + The calculated value 425 and the measured value 425 of (c).
Example 64: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (64)
Figure BDA0003762336660002022
Figure BDA0003762336660002031
1. At N 2 EtMgBr (42mL, 126mmol,3M in ether) was then added slowly to a solution of E-3_1 (20.0 g, 63.1mmol) in THF (300 mL) at-70 ℃. After addition, the mixture was stirred at-70 ℃ for 2 hours. The mixture is saturated with NH 4 Cl (500 mL) quenched and extracted with EtOAc (3 × 500 mL). The combined organic phases were washed with brine (500 mL) and Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give E-3 \ u 2 (6.50g, 30%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.57-2.49(m,1H),2.23-1.80(m,6H),1.78-1.52(m,4H),1.50-1.02(m,17H),0.97(s,3H),0.95-0.80(m,4H),0.60(s,3H)。
2. At 15 ℃ in N 2 T-BuOK (4.19g, 37.4mmol) was added to MePPh 3 Suspension of Br (13.3g, 37.4 mmol) in THF (200 mL). The mixture was stirred at 50 ℃ for 30mins. E-3 \ u 2 (6.50g, 18.7 mmol) was added portionwise to the mixture at below 50 ℃. The mixture was stirred at 50 ℃ for 1 hour. Adding NH to the mixture 4 Cl (400 mL). The organic layer was separated and concentrated in vacuo to give the crude product, which was triturated from MeOH/water (200ml, 1. After cooling the mixture was filtered and the solid was washed with MeOH/water (2x30ml, 1) and concentrated in vacuo to give E-3_3 (5.8 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.84(s,1H),4.70(s,1H),2.06-1.97(m,1H),1.94-1.55(m,12H),1.53-1.05(m,16H),0.97(s,3H),0.95-0.85(m,3H),0.55(s,3H)。
3. At 15 ℃ in N 2 Next, 9-BBN dimer (8.19g, 33.6mmol) was added to E-3_3 (5.80g, 16)8 mmol) in THF (100 mL). The reaction mixture was stirred at 60 ℃ for 1 hour. The mixture was cooled to 15 ℃. Ethanol (7.72g, 168mmol) was added at 15 ℃. Aqueous NaOH (33.6 mL,5M, 168mmol) was added dropwise at 15 ℃. H is added dropwise at 15 DEG C 2 O 2 (16.8 mL,10.0M, 168mmol). The resulting mixture was stirred at 60 ℃ for 1 hour. The aqueous phase was extracted with EtOAc (3X 100 mL). The combined organic phases were washed with brine (2X 100 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue is separated from CH at 65 DEG C 3 OH/H 2 O =1/1 (150 mL) to give E-3_4 (2.80g, 46%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.66-3.61(m,1H),3.38-3.32(m,1H),2.03-1.56(m,4H),1.56-1.51(m,5H),1.51-1.10(m,16H),1.10-1.02(m,6H),0.97(s,3H),0.96-0.88(m,3H),0.67(s,3H)。
4. DMP (5.80g, 13.7 mmol) was added to a solution of E-3 \ u 4 (2.50g, 6.89mmol) in DCM (50 mL). Then, the reaction was stirred at 20 ℃ for 30 minutes. Saturated NaHCO was added to the reaction mixture 3 Aqueous solution (50 mL), saturated Na 2 S 2 O 3 Aqueous (30 mL) and extracted with DCM (2 × 20 mL). The combined organic layers were washed with saturated NaHCO 3 Aqueous solution (3X 10 mL) and brine (20 mL) were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give E-3_5 (2.45 g, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.60-9.55(m,1H),2.26(s,1H),1.98-1.70(m,6H),1.70-1.51(m,6H),1.51-1.00(m,12H),1.00-0.89(m,10H),1.75-0.65(m,4H)。
5. In N 2 Isobutyl magnesium bromide (33.9mL, 2M in THF, 67.9 mmol) was added to a solution of E-3 \ u 5 (2.45g, 6.79mmol) in THF (10 mL) at 0 deg.C. The mixture was stirred at 20 ℃ for 16 hours. Adding NH to the mixture 4 Cl (20 mL, saturated aqueous solution), the mixture was extracted with EtOAc (2 × 30 mL). The combined organic phases were washed with brine (20 mL) and Na 2 SO 4 Drying, vacuum concentration, and purification by flash column (0-20% EtOAc in PE) to give E3_6 (1.6g, 55%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.65-3.55(m,1H),2.01-1.85(m,3H),1.85-1.49(m,3H),1.49-1.36(m,12H),1.36-1.22(m,10H),1.22-1.02(m,9H),1.02-0.98(m,4H),0.98-0.80(m,7H),0.66(s,3H)。
6. DMP (3.12g, 7.38mmol) was added to a solution of E-3_6 (1.6g, 3.69mmol) in DCM (30 mL). The reaction was then stirred at 20 ℃ for 30 minutes. Saturated NaHCO was added to the reaction mixture 3 Aqueous solution (20 mL), saturated Na 2 S 2 O 3 Aqueous (20 mL) then extracted with DCM (2 × 20 mL). The combined organic layers were washed with saturated NaHCO 3 Aqueous solution (3X 10 mL) and brine (20 mL), washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to afford E-3_7 (1.5 g, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ2.55-2.80(m,3H),2.80-2.23(m,1H),1.98-1.81(m,2H),1.81-1.1.69(m,1H),1.69-1.25(m,16H),1.25-1.01(m,10H),1.01-0.81(m,14H),0.67(s,3H)。
7. NaBH is added at 0 DEG C 4 (255mg, 6.72mmol) was added in one portion to a solution of E-3/u 7 (1.45g, 3.36mmol) in MeOH (20 mL). After addition, the mixture was stirred at 20 ℃ for 1hr and treated with NH 4 Cl (20 mL, saturated aqueous solution) was quenched. The mixture was extracted with DCM (2 × 20 mL). The combined organic phases were washed with brine (2X 10 mL) and Na 2 SO 4 Dried, filtered, concentrated and purified by flash column (0-20% EtOAc in PE) to give E-3_8 (1.2g, 83%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.80-3.70(m,1H),3.70-3.55(m,1H),2.05-1.82(m,3H),1.82-1.55(m,4H),1.55-1.35(m,5H),1.35-1.00(m,18H),1.00-0.79(m,17H),0.66(s,3H)。
8. Benzoyl chloride (1.85g, 13.2mmol) was added to a solution of E-3/u 8 (1.15g, 2.65mmol) in pyridine (20 mL). The reaction mixture was stirred at 20 ℃ for 4 hours. The reaction mixture was poured into water (20 mL). The mixture was washed with EtOAc (2X 20 mL)) And (4) extracting. The combined organic phases were washed with saturated brine (2X 10 mL) and anhydrous Na 2 SO 4 Dried, concentrated by filtration and purified by flash column (0-10% etoac in PE) to give the mixture product (1.45 g, impure) as a solid. The mixture product (1.45 g, impure) was purified by SFC (column: AD (250mm x 50mm, 10um), gradient: 30-30% B (a =0.1% nh3/H2O, B = EtOH), flow rate: 200 mL/min) to obtain solid E-3 \ u 9 (peak 2, 470mg,33%, DE% = 100%) and solid E-3 \ u 9a (peak 1, 600mg,42%, DE% = 99.1%).
E-3_9A:
1 H NMR(400MHz,CDCl 3 )δ8.10-7.99(m,2H),7.60-7.50(m,1H),7.50-7.38(m,2H),5.15-5.05(m,1H),2.05-1.70(m,6H),1.70-1.35(m,5H),1.35-1.05(m,19H),1.05-0.82(m,17H),0.65(s,3H)。
SFC Rt =3.344 min, AD _3_EtOH _DEA5 _40 _25MLin 10 min chromatography, 100% b.
E-3_9:
1 H NMR(400MHz,CDCl 3 )δ8.10-7.99(m,2H),7.60-7.50(m,1H),7.50-7.38(m,2H),5.25-5.15(m,1H),2.05-1.80(m,3H),1.80-1.45(m,15H),1.45-1.09(m,13H),1.09-0.85(m,16H),0.68(s,3H)。
SFC Rt =3.851 min, AD _3_EtOH _DEA5 _40_25ML,99.1% by 10 min chromatography.
9. NaOH (531mg, 13.3mmol) and H were added at 25 deg.C 2 O (0.5 mL) was added to a solution of E-3 \ u 9A (600mg, 1.11mmol) in THF (2 mL) and MeOH (2 mL). The solution was stirred at 50 ℃ for 48 hours. Water (10 mL) was added. The mixture was extracted with EtOAc (2 × 10 mL). The combined organic layers were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product, which was triturated with MeCN (10 mL) to give the desired product 69 (473mg, 99%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.68-3.55(m,1H),2.01-1.85(m,3H),1.85-1.70(m,1H),1.70-1.45(m,8H),1.45-1.22(m,13H),1.22-1.05(m,8H),1.05-1.86(m,15H),0.66(s,3H)。
LCMS t R =1.403 min, 2 min chromatography, 30-90AB _ELSD, purity 100.0%, MS ESI C 29 H 49 [M+H-2H 2 O] + Calculated value 397 of (a), found value 397.
Example 65: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-4-methylpentan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (65)
Figure BDA0003762336660002071
N-8-11 \ (30mg, 0.0741mmol, impure) was purified by Combi-flash (25% EtOAc in PE) to give 65 (9mg, 30%) as a solid.
65:
1 H NMR(400MHz,CDCl 3 )δ3.18-3.07(m,1H),1.98-1.81(m,2H),1.71-1.58(m,6H),1.53-1.31(m,7H),1.30-0.98(m,14H),0.97-0.78(m,14H),0.70-0.60(m,4H)。
LCMS Rt =4.387 minutes, chromatography over 7.0 minutes, 30-90AB u 7MIN, 97.6% purity, calcd for MS ESI C 27 H 45 [M+H-2H 2 O] + 369, found 369.
Example 66: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-4- (2-methylcyclopropyl) but-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (66)
Figure BDA0003762336660002081
Figure BDA0003762336660002091
1. To a solution of N-8-7_2 (1g, 2.41mmol) in THF (50 mL) at 0 deg.C was added LiAlH 4 (914mg, 24.1mmol). The grey suspension was heated at 66 ℃ for 18 hours. Cooling the reaction mixture to 0 deg.CFollowed by quenching with ice-water (914 mg), 15% w/w aqueous NaOH (914 mg), water (2.74 g). The mixture was filtered and the filter cake was washed with DCM (3 × 50 mL). The filtrate was concentrated to give a residue which was purified twice by flash chromatography (ethyl acetate 10% in PE) to give N-8-7_3 (192mg, 19%) and N-8-7_3a (3976 mg, 39%) as an oil.
N-8-7_3:
1H NMR(400MHz,CDCl3)δ5.59-5.36(m,2H),3.69-3.61(m,1H),2.25-2.12(m,1H),2.08-1.81(m,3H),1.68(d,J=10.0Hz,3H),1.64-1.54(m,9H),1.53-1.15(m,11H),1.14-0.92(m,5H),0.92-0.85(m,5H),0.83(s,4H),0.69-0.60(m,4H)。
N-8-7_3A:
1 H NMR(400MHz,CDCl 3 )δ5.62-5.37(m,2H),3.62(br d,J=10.0Hz,1H),2.20-2.06(m,1H),1.99-1.61(m,6H),1.61-1.44(m,11H),1.43-1.18(m,5H),1.16-0.94(m,6H),0.94-0.85(m,5H),0.82(s,5H),0.70-0.58(m,6H)。
2. To a solution of diethylzinc (1M in toluene, 2.59mL, 2.59mmol) in DCM (10 mL) was added CH over 15 minutes at 0 deg.C 2 I 2 (1.38g, 5.18mmol). The milky suspension was stirred at 0C for 10 minutes and a solution of preformed Charette ligand ((4R, 5R) -2- (tert-butyl) -N4, N4, N5, N5-tetramethyl-1, 3, 2-dioxaborolan-4, 5-dicarboxamide) (139mg, 0.5182mmol) and N-8-7 xu 3 (180mg, 0.4319mmol) in DCM (15 ml) was added rapidly by syringe, whereupon the reaction mixture became clear. The solution was warmed to 25 ℃ and stirred at 25 ℃ for 16h. The reaction was then quenched by addition of saturated NH 4 Aqueous Cl (150 ml) was quenched. The phases were separated and the aqueous phase was extracted with DCM (3 × 60 ml). The combined organic phases were washed with saturated NaHCO 3 Aqueous solution (150 mL), saturated Na 2 S 2 O 3 Aqueous solution (150 mL) and brine (100 mL) were washed with Na 2 SO 4 Dried, filtered and concentrated under reduced pressure and the residue purified by flash column chromatography (11% ethyl acetate in PE) to give N-8-7 \u4 (60mg, 32%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.77(br s,1H),1.99-1.87(m,2H),1.70-1.56(m,6H),1.54-1.34(m,6H),1.31-1.16(m,5H),1.15-1.05(m,1H),1.15-0.97(m,7H),0.99-0.94(m,1H),0.92-0.77(m,10H),0.69-0.58(m,6H),0.52-0.13(m,5H)。
3. To a solution of N-8-7_4A (60mg, 0.1393mmol) in pyridine (3 mL) at 25 ℃ was added benzoyl chloride (39.1mg, 0.2786mmol) followed by DMAP (6.79mg, 0.05572mmol). The reaction mixture was stirred at 60 ℃ for 16 hours. The reaction mixture was diluted with DCM (80 mL). DCM phase was purified with water (100 mL), 1.0M aqueous HCl (2X 100 mL), 10% NaHCO 3 Aqueous (2X 100 mL), brine (100 mL), na 2 SO 4 Dried, filtered and concentrated in vacuo to give an oil which was purified by flash column (1% ethyl acetate in PE) to give N-8-7 \u5 (24mg, 32%) as an oil.
LCMS Rt =1.431 min, chromatography over 2 min, 5-95AB _220&254, purity 90%, MS ESI C 36 H 53 O 2 [M-H 2 O+H] + Calcd for 517.3, found 517.3.
SFC peak 1: rt =5.703 min, with 10 min chromatography, AD _3_etoh _dea _5_40_25ml ("Chiralpak AD-3 × 4.6mm i.d.,3um mobile phase: A: CO 2B: ethanol (0.05% DEA) gradient 5% to 40% B in 5 minutes and held at 40% for 2.5 minutes, then 5% B was held for 2.5 minutes at a flow rate of 2.5mL/min and a column temperature of 35 ℃').
N-8-7-u 5 (24mg, 0.04487mmol) was purified by SFC (column: AD (250mm. 30mm,5 um)), gradient: 40-40% B (A =0.1% NH) 3 /H 2 O, B = EtOH), flow rate: 50 mL/min) to yield impure N-8-7/u 6 (RT: 5.732 19mg, impure) as a solid. The isomer was not obtained.
LCMS Rt =1.435 min, chromatography over 2 min, 5-95AB _220&254, purity 98%, MS ESI C 36 H 53 O 2 [M-H 2 O+H] + Calcd for 517.3, found 517.3.
SFC Rt =5.732 min, AD _ 3. Mu. EtOH _DEA5. Mu. 40. Mu. 25ML,97.76% by 10 min chromatography.
5. To a solution of N-8-7 (19mg, 0.0355mmol) in THF/MeOHTo the solution in (0.5 mL/0.5 mL) was added a solution of KOH (19.8 mg, 0.0.3552mmol) in water (0.2 mL). The reaction mixture was stirred at 55 ℃ for 16 hours. To this mixture was added HCl (0.2M, 50mL). The suspension was extracted with DCM (2 × 60 mL). The combined organic phases were then treated with 3% NaHCO 3 Aqueous solution (80 mL) and brine (80 mL) were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solid which was purified by flash chromatography (ethyl acetate in PE, 15%) to give 66 (2mg, 13%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.77(br s,1H),1.98-1.87(m,2H),1.70-1.57(m,7H),1.54-1.28(m,11H),1.26-0.94(m,12H),0.91-0.84(m,7H),0.83(s,3H),0.72-0.60(m,4H),0.51-0.15(m,3H)。
LCMS Rt =1.315 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, MS ESI C 29 H 49 O[M-H 2 O+H] + 395.3, found 395.3.
Example 67: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -4-cyclopentyl-3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (67)
Figure BDA0003762336660002121
1. Pd (OH) 2 (160 mg, anhydrous) was added to a solution of 32 (80mg, 0.18mmol) in MeOH (20 mL). The mixture was heated at 50 ℃ in H 2 Stirring was carried out for 48 hours under (50 Psi). The mixture was filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 67 (10 mg, 12%) and 84 (30mg, 37%) as solids.
67:
1 H NMR(400MHz,CDCl 3 )δ3.76-3.66(m,1H),2.01-1.78(m,5H),1.76-1.58(m,7H),1.52-1.31(m,13H),1.28-1.10(m,10H),1.09-0.99(m,4H),0.96(s,3H),0.93-0.86(m,6H),0.67(s,3H)。
LCMS Rt =1.508 min at 2.0 min colorSpectrometry, 30-90AB _E, purity 100%, MS ESI C 30 H 49 [M+H-2H 2 O] + The calculated value 409 of (1) is actually measured value 409.
Example 68: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxyhept-5-yn-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (68)
Figure BDA0003762336660002131
1. Crude N-8-7 \/2B (250mg, 0.868mmol) was further purified by SFC (column: AD (250mm. Multidot. 30mm, 10um)), gradient: 35-35% of B (A =0.1% 3 /H 2 O, B = EtOH), flow rate: 60 mL/min) to give 41 as a solid (peak 2, 81mg, 33%) and 68 as a solid (peak 1, 78mg, 31%).
68:
1 H NMR(400MHz,CDCl 3 )δ3.87-3.78(m,1H),2.21-2.12(m,1H),1.99-1.86(m,2H),1.80(s,3H),1.73-1.51(m,8H),1.51-1.42(m,4H),1.42-1.20(m,8H),1.20-0.95(m,7H),0.95-0.79(m,8H),0.95(s,4H)。
LCMS Rt =1.188 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, MS ESI C 28 H 45 O[M+H-H 2 O] + Calculated value 397 of (a), found value 397.
SFC Rt =6.465 minutes, AD _3_EtOH _DEA5 _40 _25MLin 10 minute chromatography, 100% w/w.
Example 69: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -17- ((2S, 3S) -5-cyclopropyl-3-hydroxypentan-2-yl) -3-ethyl-10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (69)
Figure BDA0003762336660002141
1. NaBH is reacted at 15 ℃ 4 (2.46g, 65.1mmol) was added portionwise to a solution of S-500-6-19-u 3 (700mg, 1.63mmol) in THF (5 mL) and MeOH (5 mL). In that After stirring for 20 minutes at 15 ℃ the mixture is taken up in NH 4 Cl (20 mL, saturated aqueous) was quenched and extracted with EtOAc (50 mL). The organic layer was separated and concentrated in vacuo to give 760mg of the mixture as a solid, which was separated by flash column (0-35% DCM/EtOAc (1/1) in PE) to give 69 (330mg, 47%) and 6 (250mg, 35%, impure) as solids. Impure 6 (250 mg) was further separated by flash column (0-35% DCM/EtOAc (1/1) in PE) to give 6 (170mg, 23%) as a solid.
69:
1 H NMR(400MHz,CDCl 3 )δ5.33-5.23(m,1H),3.75-3.63(m,1H),2.41-2.31(m,1H),2.09-1.85(m,4H),1.78-1.59(m,5H),1.53-1.38(m,9H),1.38-1.05(m,9H),1.03(s,3H),1.00-0.91(m,1H),0.91(d,J=6.4Hz,3H)0.85(t,J=7.6Hz,3H),0.69(s,3H),0.68-0.60(m,1H),0.45-0.36(m,2H),0.09--0.08(m,2H)。
LCMS Rt =1.387 min, chromatography at 2.0 min, 30-90 u AB _E, 98.1% purity, MS ESI C 29 H 47 O[M+H-H 2 O] + The calculated value 411 of (a), the measured value 411.
S-500-6-19:
1 H NMR(400MHz,CDCl 3 )δ5.32-5.24(m,1H),3.77-3.66(m,1H),2.41-2.31(m,1H),2.09-1.91(m,3H),1.79-1.59(m,6H),1.55-1.21(m,14H),1.21-1.06(m,4H),1.03(s,3H),1.00-0.95(m,1H),0.93(d,J=6.8Hz,3H)0.85(t,J=7.6Hz,3H),0.70(s,3H),0.68-0.62(m,1H),0.49-0.38(m,2H),0.11-0.02(m,2H)。
LCMS Rt =1.380 min, chromatography at 2.0 min, 30-90_AB _E, purity 100%, MS ESI C 29 H 47 O[M+H-H 2 O] + The calculated value 411 of (a), the measured value 411.
Example 70: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-ethyl-17- ((2S, 3S) -3-hydroxy-4- (3-methyloxetan-3-yl) but-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (70).
Figure BDA0003762336660002151
1. At N 2 Adding Mg (807mg, 33.2mmol) and I at 50-55 deg.C 2 (1 mg) to a suspension in THF (2 mL) was added dropwise a solution of N-014-005 (2.5g, 15.1mmol) in THF (8 mL). The mixture was stirred at 55 ℃ for 1h. The mixture was diluted with THF (10 mL) and used directly in the next step without monitoring. To a solution of N-14-12 (1.01g, 2.83mmol) in THF (10 mL) at 0 deg.C was added the freshly prepared 3- [ (magnesium bromide) methyl group ]-3-methyloxetane (15 mmol in 20mL THF). The mixture was stirred at 15 ℃ for 4h. Adding NH to the mixture 4 Cl (20mL, 10. Aq.). The mixture was extracted with EtOAc (30 mL). The organic layer was separated and concentrated in vacuo. The residue was purified by flash column (0-30% EtOAc in PE) to give a mixture (190mg, 15%) as a white solid, which was purified by SFC (column: AD (250mm. Multidot. 30mm,5 um), conditions: 0.1% NH3H2O ETOH, gradient: 50% to 50%, flow rate (mL/min): 60mL/min,25 ℃) to give 33 (peak 1, 110mg, 9%) and 70 (peak 2, 30mg, impure) as white solids. Impure 70 (30 mg, impure) was purified by column chromatography on silica gel (15% etoac in PE) to give 70 (10mg, 5%) as a white solid.
33:
1 H NMR(400MHz,CDCl 3 )δ5.30-5.26(m,1H),4.59-4.70(m,1H),4.50-4.48(m,1H),4.36-4.33(m,1H),3.83(s,1H),2.40-2.33(m,1H),2.10-1.50(m,17H),1.49-1.35(m,9H),1.30-0.80(m,13H),0.68(s,3H)。
LCMS Rt =1.069 min, 3 min chromatography, 30-90AB _2MIN _E.M, 100% purity, MS ESI C 29 H 49 O 3 [M+H] + Calculated value 445, found value 445.
70:
1 H NMR(400MHz,CDCl 3 )δ5.30-5.26(m,1H),4.10-4.02(m,1H),3.81-3.70(m,1H),3.50-3.41(m,2H),3.34-3.30(m,1H),2.35-2.31(m,1H),2.10-1.50(m,18H),1.49-1.05(m,13H),1.05-0.90(m,4H),0.90-0.80(m,3H),0.67(s,3H)。
LCMS Rt =1.115 min, chromatography over 3 min, 30-90AB _2MIN _E.M, 100% purity, MS ESI C 29 H 49 O 3 [M+H] + Calcd value 445, found value 445.
Example 71: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3- (methoxymethyl) -10, 13-dimethyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybut-2-yl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (71)
Figure BDA0003762336660002161
1. In N 2 TMSCF was added to a solution of N-004-022_5 (200mg, 0.531mmol), csF (40.2mg, 0.265mmol) in THF (5 mL) at 0 deg.C 3 (187mg, 1.32mmol). The mixture was stirred at 25 ℃ for 1 hour. To the mixture was added TBAF.3H2O (836 mg, 2.65mmol). After stirring for 2hrs at 25 ℃, the mixture was 50% 4 Cl (20 mL) quenched and extracted with EtOAc (2 × 10 mL). The combined organic phases were washed with brine (20 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, PE/EA = 10/1) to give 9 (56mg, 24%) and 71 (30 mg, impure) as white solids.
71 (30mg, 0.067mmol) was recrystallised from n-hexane (2 mL) at 25 ℃ to give 71 (24mg, 10%) as a white solid.
71:
1 H NMR(400MHz,CDCl 3 )δ4.08-4.00(m,1H),3.39(s,3H),3.24-3.18(m,2H),2.22-2.15(m,1H),2.02-1.77(m,5H),1.75-1.68(m,2H),1.64-1.52(m,5H),1.47-1.31(m,6H),1.28-1.01(m,10H),0.97(s,3H),0.67(s,3H)。
LCMS Rt =1.105 min, chromatography over 2 min, 30-90AB _POS _E.M, 100% purity, MS ESI C 25 H 41 F 3 O 3 [M+Na] + Calculated value of 469, found 469.
Example 72: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-ethyl-17- ((2S, 3S) -3-hydroxyhex-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (72)
Figure BDA0003762336660002171
1. Propylmagnesium bromide (3.34mL, 6.69mmol,2M in THF) was added slowly to a solution of S-500-6-1 \u1 (800mg, 2.23mmol) in THF (30 mL) at 0 ℃. After the addition, the mixture was stirred at 15 ℃ for 1 hour. Mixing the mixture with sat 4 Cl (40 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases were washed with brine (2 × 30 mL) and Na 2 SO 4 Dried, filtered and concentrated and purified by combi-flash (0-15% etoac in PE) to give 72 (500mg, 56%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.31-5.26(m,1H),3.72-3.64(m,1H),2.41-2.31(m,1H),2.07-1.85(m,4H),1.77-1.69(m,1H),1.62-1.54(m,3H),1.52-1.38(m,9H),1.37-1.16(m,6H),1.15-1.01(m,7H),0.99-0.88(m,7H),0.87-0.82(m,3H),0.68(s,3H)。
LCMS Rt =4.979 min, chromatography at 7.0 min, 30-90AB _E, 98.8% purity, MS ESI C 27 H 43 [M+H-2H 2 O] + The calculated value 367 and the actual value 367 of (a).
Example 73: synthesis of (3S, 5S,8R,9R,10S,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -3, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (73).
Figure BDA0003762336660002181
Figure BDA0003762336660002191
1. At N 2 A solution of G-21-1 (40g, 110mmol) in anhydrous dioxane (1L) was added downwardSodium methoxide (29.7g, 550mmol) was added thereto. The mixture was stirred at 110 ℃ for 16 hours. TLC showed almost complete consumption of starting material. The solvent was removed to 1/3 volume and the mixture was acidified with 2M HCl to pH = 5-6, extracted with DCM (500ml × 3), washed with aqueous sodium bicarbonate (500 mL) and brine (500 mL), dried over sodium sulfate and concentrated. The residue was purified by column chromatography on silica gel (PE: EA: meOH =3, 0.1) to give G-21-4A (11g, 33.1%) as a white solid.
2. A solution of Li (2.54g, 363mmol) was added portionwise to liquid ammonia (1000 mL, prepared over 1.5 hours in 13-601) at-70 ℃. The mixture was stirred at-70 ℃ for 30 minutes until all Li was dissolved. A solution of G-21-4A (11g, 36.3mmol) and tert-BuOH (5.38g, 72.6 mmol) in 400ml of anhydrous tetrahydrofuran was added dropwise and stirred for 90mins until the reaction mixture became pale yellow. TLC (PE: EA =1, pma) showed most of the STM was consumed. Ammonium chloride (15 g) was added and the excess ammonia was evaporated on standing. The residue was extracted with 0.5N HCl (500 mL) and dichloromethane (500mL. Times.2). The combined organic layers were washed with saturated NaHCO 3 Washing with Na 2 SO 4 Dried, filtered and concentrated to give a mixture of G-21-5A and G-21-5B (10G, impure), which was used directly in the next step without further purification.
3. To a solution of G-21-5A and G-21-5B (10g, 27.9mmol) in 100mL of anhydrous dichloromethane were added PCC (16.6g, 65.6mmol) and silica gel (16.6G). After stirring for 2h at 25 ℃, TLC (PE: EA =1, pma) showed depletion of STM. The resulting solution was concentrated and purified by chromatography on silica gel (petroleum ether/ethyl acetate =5:1 to 2) to give G-21-6A (4.6G, 46.4%) as a white solid.
4. Trimethylaluminum (2M in toluene, 39.5mL, 79.1mmol) was added dropwise to a solution of BHT (34.8g, 158mmol) in toluene (120 mL) at 0 deg.C under nitrogen. After stirring at 20 ℃ for 30 minutes, a solution of G-21-6A (8g, 26.4mmol) in toluene (80 mL) was added dropwise at-70 ℃ under nitrogen. After stirring at-70 ℃ for 30 min, meMgBr (3M in ether, 26.3mL,79.1mmol,3M in ether) was added dropwise. The resulting mixture was stirred at-70 ℃ for 1 hour, poured slowly into ice-cooled aqueous citric acid (300 mL) and extracted with EtOAc (3 × 100 mL). The combined organic layers were washed with brine (300 mL), dried over anhydrous sodium sulfate, filtered, concentrated and purified by combi-flash (0-40% etoac in PE) to give N-4-10 \u1 (6.5g, 77%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ2.60-2.49(m,1H),2.47-2.37(m,2H),2.34-2.19(m,3H),2.14-2.03(m,1H),2.00-1.82(m,3H),1.73-1.58(m,3H),1.56-1.46(m,2H),1.36-1.26(m,3H),1.24(s,3H),1.21-1.07(m,2H),1.04(s,3H),1.00-0.84(m,1H),0.82(s,3H)。
5. At N 2 To a suspension of bromo (ethyl) triphenylphosphorane (22.6 g, 61.1mmol) in anhydrous THF (200 mL) was added potassium 2-methylpropan-2-olate (6.84g, 61.1mmol) at 20 ℃. After stirring at 40 ℃ for 30 minutes, a solution of N-4-10_1 (6.5g, 20.4 mmol) in dry THF (50 mL) was added slowly. The resulting mixture was stirred at 40 ℃ for 10 minutes and then with NH 4 Aqueous Cl (400 mL) was quenched and extracted with EtOAc (2 × 150 mL). The combined organic phases were dried over sodium sulfate, filtered, concentrated and purified by column chromatography on silica gel (0-25% etoac in PE) to give N-4-10 \u2 (5.5g, 82%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ5.22-5.13(m,1H),2.91-2.81(m,1H),2.62-2.51(m,1H),2.50-2.39(m,2H),2.38-2.24(m,1H),1.91-1.81(m,1H),1.80-1.70(m,4H),1.55-1.41(m,4H),1.36-1.25(m,5H),1.23(s,3H),1.21-1.04(m,3H),1.01(s,3H),0.98-0.84(m,2H),0.81(s,3H)。
6. At 15 ℃ in N 2 To a mixture of N-4-10_2 (5.5g, 16.6 mmol) in THF (100 mL) was added 9-BBN dimer (8.10g, 33.2mmol). After stirring at 50 ℃ for 1 hour, the mixture was cooled to 15 ℃. Aqueous NaOH (33.2mL, 5M, 166mmol) was added dropwise at below 15 deg.C, followed by H addition at below 15 deg.C 2 O 2 (18.8 g,30%,166 mmol). The mixture was extracted with EtOAc (3 × 100 mL). Sat. Na for combined organic phases 2 S 2 O 3 (5X 100 mL) and washed with Na 2 SO 4 Dried, filtered and concentrated to give 7g of crude material, which was used directly in the next step.
7. To a solution of N-4-10 \u3 (7g, 19.9mmol) in DCM (300 mL) was added DMP (25.2g, 59.6 mmol). After stirring for 10 min at 20 ℃ the reaction mixture was saturated with NaHCO 3 The solution (500 mL) was quenched until the pH of the aqueous layer became about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (200 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3 × 400 mL), sat 3 (400 mL), brine (400 mL), na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-20% EtOAc in DCM) to give N-4-10 \u4 (4 g, 58%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ2.77-2.67(m,1H),2.65-2.38(m,3H),2.32-2.17(m,1H),2.09(s,3H),1.88-1.63(m,7H),1.59-1.49(m,3H),1.35-1.21(m,7H),1.19-1.09(m,2H),1.01(s,3H),0.96-0.84(m,1H),0.57(s,3H)。
8. To MePh 3 To a suspension of PBr (8.18g, 23.0mmol) in THF (100 mL) was added t-BuOK (2.57g, 23.0mmol). After stirring at 40 ℃ for 10 minutes, the mixture was slowly added dropwise to a solution of N-4-10/-4 (4 g,11.5 mmol) in THF (50 mL) at 20 ℃. After addition, the mixture is washed with NH 4 Cl (200 mL) quenched and extracted with EtOAc (3 × 80 mL). The combined organic phases are treated with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-25% etoac in PE) to give N-4-10 \u5 (3.2g, 80%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ4.88(s,1H),4.70(s,1H),2.48-2.37(m,2H),2.31-2.22(m,2H),1.89-1.77(m,4H),1.75-1.61(m,7H),1.54-1.45(m,2H),1.34-1.29(m,2H),1.28-1.24(m,3H),1.23(s,3H),1.17-1.05(m,2H),1.01(s,3H),0.93-0.83(m,1H),0.51(s,3H)。
9. At 15 ℃ in N 2 To a mixture of N-4-10_5 (3.2g, 9.28mmol) in THF (100 mL) was added 9-BBN dimer (4.51g, 18.5 mmol) next. After stirring at 50 ℃ for 1 hour, the mixture was cooled to 15 ℃. Aqueous NaOH (18.5mL, 5M, 92.8mmol) was added dropwise at below 15 ℃ followed by H addition at below 15 ℃ 2 O 2 (10.5g, 30%,92.8 mmol). The mixture was extracted with EtOAc (3 × 100 mL). Sat. Na for combined organic phases 2 S 2 O 3 (5X 100 mL) and washed with Na 2 SO 4 Dried, filtered and concentrated to give 5g of crude material, which was used directly in the next step.
10. To a solution of N-4-10 \ u 5A (5 g,13.7 mmol) in DCM (300 mL) was added DMP (11.6 g,27.4 mmol). After stirring for 10 min at 20 ℃ the reaction mixture was taken up with saturated NaHCO 3 The solution (300 mL) was quenched until the pH of the aqueous layer became about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (100 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3 × 300 mL), sat 3 (300 mL), brine (300 mL), washed with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-10% acetone in DCM) to give N-4-10_7 (1g, 20%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ9.58-9.55(m,1H),2.52-2.27(m,4H),2.08-1.96(m,1H),1.84-1.62(m,8H),1.51-1.39(m,3H),1.32-1.21(m,7H),1.17-1.06(m,5H),1.01(s,3H),0.94-0.83(m,1H),0.66(s,3H)。
11. In N 2 Isopentylmagnesium bromide (1.65mL, 3.30mmol,2M in ether) was added dropwise at 0 ℃ to a solution of N-4-10 \/6 (400mg, 0.832mmol) in THF (20 mL). After stirring at 0 ℃ for 10 minutes, the mixture was washed with sat 4 Cl (60 mL) quenched and extracted with EtOAc (2 × 30 mL). The combined organic phases were washed with brine (60 mL) and Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-10% acetone in DCM) to give 73 (200mg, 42%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ3.65-3.56(m,1H),2.55-2.49(m,1H),2.46-2.38(m,1H),2.32-2.25(m,1H),2.10-1.98(m,1H),1.83-1.62(m,7H),1.57-1.44(m,4H),1.42-1.25(m,7H),1.24-1.20(m,4H),1.19-1.04(m,5H),1.01(s,3H),0.94-0.82(m,10H),.0.63(s,3H)。
LCMS Rt =3.381 minutes, to7.0 min chromatography, 30-90AB _E, 100% purity, MS ESI C 28 H 47 O 2 [M+H-H 2 O] + Calculated value 415, found value 415.
Example 74: synthesis of (3S, 5S,8R,9R,10S,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -3, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (74)
Figure BDA0003762336660002231
Figure BDA0003762336660002241
1. In N 2 t-BuOH (1.7L) was added to a three-neck round bottom flask at 35 ℃ and stirred for 10mins. t-BuOK (292g, 2.61mol) was added to the mixture and stirred until the reaction became clear. Then, S-310-B9-1 (65g, 238mmol) was added to the above mixture and heated at 35 ℃ under N 2 Stirring for 1.5h. The reaction mixture was poured into 10% aqueous acetic acid (2L) and stirred for 30mins, during which time the temperature was kept below 10 ℃. The mixture was then treated with water (1.5L) and the pH was adjusted with NaHCO 3 Adjusted to 7-8 and the mixture is stirred for 30mins. The aqueous phase was extracted with MTBE (3L). The organic layer was separated, washed with brine (3X 1L), and dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated below 35 ℃ to give S-200-N19-3_1 (65 g, crude) as an oil. The crude residue was used directly in the next step.
2. To a solution of 2, 6-di-tert-butyl-4-methylphenol (340g, 1.54mol) in toluene (700 mL) was added AlMe dropwise at 0 deg.C 3 (385mL, 770mmol,2M in toluene). The mixture was stirred at 25 ℃ for 1hr and used directly as MAD solution. In N 2 A solution of 200-N19-3_1 (60g, 220mmol) in dry toluene (200 mL) and dry DCM (200 mL) was added to the MAD solution over 30mins at-70 ℃. The reaction mixture was stirred at-70 ℃ for 1h. MeMgBr (220mL, 660mmol,3M in ether) was then added dropwise at-70 ℃ and stirred for 1h. The reaction was poured into saturated aqueous citric acid (2L) at 0 deg.C and stirred for 30min with EtOAc (2X 1L)And (4) extracting. The combined organic phases were washed with saturated brine (2X 1L) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE/EtOAc =10/1 to 5/1) to give 200-N19-M22 — 1 (33g, 52%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.46-5.42(m,1H),2.25-2.40(m,1H),2.21-1.60(m,13H),1.35-1.21(m,4H),1.13(s,3H),0.98-0.83(m,6H)。
3. At 25 ℃ in N 2 Next, t-BuOK (31.0g, 277mmol) was added to Ph in one portion 3 A suspension of PEtBr (102g, 277mmol) in dry THF (500 mL). After stirring for 30 minutes at 25 deg.C, 200-N19-M22-1 (20g, 69.3mmol) was added and stirred for 2h at 25 deg.C. The reaction was carried out at 0 ℃ with aq 4 Quenched with Cl (800 mL) and extracted with EtOAc (2X 500 mL). The combined organic phases were washed with brine (2X 500 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE/EtOAc =10/1 to 5/1) to give N-4-14_1 (15g, 72%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.43-5.40(m,1H),5.16-5.10(m,1H),2.41-2.33(m,1H),2.28-1.86(m,8H),1.78-1.71(m,1H),1.69-1.50(m,11H),1.41-1.10(m,6H),0.94-0.81(s,3H)。
4. 9-BNN dimer (66.9g, 299mmol) was added to a solution of N-4-14 (1) (30g, 99.8mmol) in anhydrous THF (500 mL) and at 0 deg.C in N 2 Stirring for 30mins. The reaction mixture was warmed to 50 ℃ and stirred for 1 hour. After cooling to 0 ℃ EtOH (100 mL) was added. NaOH.aq (99.8mL, 5M, 499mmol) was added very slowly. Slow addition of H 2 O 2 (53.0 g,499mmol,30% in water) and the internal temperature is kept below 30 ℃. The mixture was warmed to 50 ℃ and stirred for 1 hour. The reaction mixture was cooled and ice-water (1L) was added and stirred for 30 minutes. Filtered and concentrated in vacuo to afford N-4-11 \u2 (30 g, crude) as a solid. The crude residue was used directly in the next step.
5. Silica gel (150 g) and PCC (81.0 g, 376mmol) were added to a solution of N-4-14_2 (30 g, crude material) in DCM (500 mL). The reaction mixture was warmed to 40 ℃ and stirred for 1 hour. The reaction mixture was cooled, treated with PE (500 mL), filtered through a pad of silica gel and the solid washed with PE/DCM (500/500 mL). The mother liquor was filtered and concentrated in vacuo to give the crude product. The residue was purified by silica gel chromatography (PE/EtOAc =8/1 to 5/1) to give N-4-14_3 (20 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.43-5.40(m,1H),2.57-2.50(m,1H),2.21-2.08(m,6H),1.77-1.43(m,10H),1.37-1.12(m,9H),1.00-0.82(m,2H),0.64(s,3H)。
6. At 0 ℃ in N 2 Next, t-BuOK (14.1g, 126mmol) was added to Ph in one portion 3 PMeBr (44.8 g, 126mmol) in dry THF (300 mL). The reaction mixture was stirred at 25 ℃ for 30 minutes. N-4-14 (20g, 63.1mmol) was added. The reaction mixture was warmed to 40 ℃ and stirred for 1 hour. The reaction was poured into ice-water (500 mL) at 0 ℃. The aqueous phase was extracted with EtOAc (2 × 400 mL). The combined organic phases were washed with brine (2X 300 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel column (PE/EtOAc =8/1 to 5/1) to give N-4-14_4 (19 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.43-5.40(m,1H),4.85(s,1H),4.71(s,1H),2.23-2.13(m,2H),1.87-1.64(m,11H),1.42-1.40(m,2H),1.29-1.08(m,8H),0.97-0.80(m,3H),0.59(s,3H)。
7. At 0 ℃ in N 2 Next, 9-BNN dimer (40.5g, 181mmol) was added in one portion to a solution of N-4-14_4 (19g, 60.4mmol) in dry THF (300 mL). The mixture was warmed to 50 ℃ and stirred for 1h. The reaction mixture was cooled and EtOH (100 mL) was added. NaOH aq. (60.3mL, 5M, 302mmol) was added very slowly. Slow addition of H 2 O 2 (34.0 g,302mmol,30% in water) and the internal temperature was kept below 10 ℃. The mixture was warmed to 50 ℃ and stirred for 1 hour. After cooling, ice-water (1L) was added and stirred for 30 minutes. The precipitated solid was filtered off. The filter cake was air dried to give N-4-11-u 5 (17 g, crude) as a solid, which was used directly in the next step.
8. To a solution of N-4-14 (17 g, crude) in DCM (300 mL) at 25 deg.C was added silica gel (60 g) and PCC (43.9g, 204mmol) in one portion. The reaction mixture was warmed to 40 ℃ and stirred for 1 hour. The reaction mixture was cooled and PE (200 mL) was added. The mixture was filtered through a pad of silica gel and the solid was washed with PE/DCM (200/200 mL). Filtered and concentrated in vacuo to give a solid. The residue was purified by silica gel chromatography (PE/EtOAc =8/1 to 5/1) to give N-4-14 \u6 (5.5 g, impure) as a solid. The residue was recrystallized from MeCN (50 mL) at 82 ℃ to give N-4-14 \u6 (5g, 91%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.60-9.56(m,1H),5.42-5.40(m,1H),2.58-2.51(m,1H),2.40-1.85(m,9H),1.44-1.04(m,16H),1.00-0.80(m,3H),0.75-0.71(m,3H)。
9. In N 2 Mg (3.96g, 165mmol) and I were added at 25 ℃ to 2 (1 mg) to a suspension in anhydrous THF (20 mL) was added dropwise a solution of 1 (12.5g, 82.7 mmol) in anhydrous THF (63 mL) and the internal temperature was raised to 65 ℃ and stirred for 2 h. The mixture was used directly in the next step. At 0 ℃ in N 2 Isopentylmagnesium bromide (83.0mL, 1M in THF) was added all at once to a solution of N-4-14-u 6 (5g, 15.1mmol) in anhydrous THF (50 mL). The reaction mixture was warmed to 15 ℃ and stirred for 1 hour. Adding saturated NH to the reaction mixture 4 Aqueous Cl (100 mL). The aqueous phase was extracted with EtOAc (3X 100 mL). The combined organic phases were washed with saturated brine (2X 200 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product. The crude product was purified by silica gel column (0-20% etoac in PE) to give N-4-14 \u7 (2.5 g, impure) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.40-5.37(m,1H),3.67-3.61(m,1H),2.19-1.71(m,9H),1.64-1.28(m,13H),1.18-1.03(m,7H),0.95-0.78(m,12H),0.69(s,3H)。
10. To a solution of DMP (10.5g, 24.8mmol) at 25 ℃ was added a solution of N-4-14 \7 (2.5g, 6.20mmol) in DCM (40 mL). The reaction mixture was warmed to 40 ℃ and stirred for 1h. The reaction mixture was saturated with NaHCO 3 Dissolving in waterThe solution is quenched at a pH of 7-8 below 10 ℃. The suspension was filtered. The DCM phase in the filtrate was separated and saturated NaHCO was used 3 /Na 2 S 2 O 3 Aqueous solution (1, 2x30ml) and the combined organic phases were washed with saturated brine (2 x30 mL) and Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solid. The residue was purified by flash column (0-30% etoac in PE) to give N-4-14 \u7o (1.5g, 60%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.41-5.39(m,1H),2.56-2.31(m,3H),2.19-1.83(m,5H),1.84-1.42(m,12H),1.30-0.97(m,12H),0.96-0.77(m,8H),0.74-0.70(m,3H)。
11. To a solution of N-4-14 u 7O (1.5g, 3.74) in MeOH (10 mL) at 25 deg.C was slowly added NaBH 4 (1.42g, 37.4 mmol) and stirred for 2 hours. Ice-water (100 mL) was added and the mixture was stirred for 30mins. The aqueous phase was extracted with DCM (2 × 20 mL). The combined organic phases were washed with saturated brine (2X 20 mL) and anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solid. The residue was purified by silica gel chromatography (PE/EtOAc =8/1 to 5/1) to give N-4-14 \u7 (1g, 67%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.41-5.39(m,1H),3.65-3.63(m,1H),2.20-2.16(m,1H),2.11-1.88(m,5H),1.86-1.54(m,10H),1.33-0.99(m,14H),0.95-0.79(m,11H),0.70(s,3H)。
SFC peak 1: rt =4.644 min and peak 2rt =5.240 min, AD _3 \ u etoh _dea5 _40_25ml ("" column: chiralpak AD-3 × 4.6mm i.d.,3um mobile phase: a: CO 2B: ethanol (0.05% dea) gradient: 5% to 40B in 5 min and held at 40% for 2.5 min, then 5% B held for 2.5 min, flow rate: 2.5mL/min, column temperature: 35 ℃ "") in 10 min chromatography.
12. N-4-14 \u7 (1g, 2.48mmol) was purified by SFC (column: AD (250mm. 30mm, 5um), condition: 0.1% NH 3 H 2 O ETOH, start: b:40%, end B: 40%) to give 80 (Peak 2, 300mg, impure) and 83 (Peak 1, 250mg, impure) as solids. 80 (300 mg, impure) was weighed at 82 ℃ from MeCN (4 mL) The crystals were refluxed for 1 hour. The stirred mixture was cooled to 25 ℃. The suspension was vacuum filtered to give 80 (150mg, 15%) as a solid. 83 (250 mg, impure) was recrystallized from MeCN (3 mL) at reflux for 1 h at 82 ℃. The stirred mixture was cooled to 25 ℃. The suspension was vacuum filtered to provide 83 (150mg, 15%) as a solid.
83:
1 H NMR(400MHz,CDCl 3 )δ5.41-5.39(m,1H),3.62-3.60(m,1H),2.22-1.89(m,6H),1.64-1.49(m,9H),1.46-1.11(m,16H),0.98-0.86(m,10H),0.70(s,3H)。
LCMS Rt =1.268 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 27 H 42 [M+H-2H 2 O] + The calculated value 367 and the measured value 367 of (4).
SFC Rt =4.609 min, AD _3_EtOH _DEA5 _40 _25MLin 10 min chromatography, 100% b.
80:
1 H NMR(400MHz,CDCl 3 )δ5.41-5.39(m,1H),3.63-3.62(m,1H),2.22-1.67(m,10H),1.64-1.36(m,12H),1.16-1.03(m,8H),0.98-0.80(m,11H),0.70(s,3H)。
LCMS Rt =1.276 min, chromatography at 2.0 min, 30-90AB, 99% purity, MS ESI C 27 H 42 [M+H-2H 2 O] + The calculated value 367 and the measured value 367 of (4).
SFC Rt =5.236 min, AD _3_EtOH _DEA5 _40 _25MLin 10 min chromatography, 100% b.
13. Dry Pd (OH) under Ar 2 (200 mg) was added to a solution of 80 (150mg, 0.372mmol) in THF (3 mL) and MeOH (3 mL). The suspension was degassed under vacuum and washed with H 2 The purge was performed three times. Mixing the mixture in H 2 Stirring was carried out at 50 ℃ for 12h (50 psi) to give a black suspension. The reaction mixture was filtered through a pad of celite and washed with DCM (3 × 50 mL). The filtrate was concentrated in vacuo to afford an oil. The residue was purified by silica gel chromatography (PE/EtOAc =8/1 to 5/1) to give 74 (20mg, 13%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.63-3.61(m,1H),1.96-1.85(m,2H),1.78-1.50(m,8H),1.45-1.19(s,12H),1.17-1.00(m,11H),0.98-0.83(m,11H),0.72-0.59(m,3H)。
LCMS Rt =1.333 min, chromatography at 2.0 min, 30-90AB, 99% purity, MS ESI C 27 H 44 [M+H-2H 2 O] + Found 369, found 369.
Example 75: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -3,10, 13-trimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (75)
Figure BDA0003762336660002301
1. DMP (2.44g, 5.76mmol) was added to a solution of 15_3a (1g, 2.88mmol) in DCM (10 mL). The reaction was then stirred at 25 ℃ for 10 minutes. The reaction mixture was purified by addition of saturated NaHCO 3 Aqueous solution (20 mL) solution and saturated Na 2 S 2 O 3 Aqueous (20 mL) solution was quenched and extracted with DCM (2 × 50 mL). The combined organic layers were washed with saturated NaHCO 3 Aqueous (3 × 50 mL) solution and brine (50 mL) were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give S-500-2-9_1 (1 g, crude material) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.57(br.s,1H),5.35-5.25(m,1H),2.50-2.30(m,2H),2.05-1.95(m,3H),1.95-1.80(m,1H),1.75-1.65(m,1H),1.65-1.60(m,3H),1.55-1.50(m,2H),1.50-1.40(m,2H),1.40-1.30(m,1H),1.25-1.20(m,2H),1.20-1.15(m,2H),1.15-1.10(m,6H),1.05-0.95(m,5H),0.90-0.70(m,1H),0.68(s,3H)。
2. Magnesium (641mg, 26.4mmol) and I 2 (33.5mg, 0.132mmol) of the mixture was stirred at 60 ℃ and N 2 A solution of isoamyl magnesium bromide (2g, 13.2mmol) in THF (20 mL) was added dropwise. The reaction mixture was then stirred at 60 ℃ for 1h. The reaction mixture was used directly as an isopentyl magnesium bromide solution without any use ofAnd (5) purifying. At 0 ℃ in N 2 Next, the Grignard solution was added to a solution of S-500-2-9-u 1 (1g, 2.90mmol) in THF (10 mL). The reaction mixture was then stirred at 25 ℃ for 1h. Adding saturated NH to the reaction mixture 4 Aqueous Cl (50 mL), extracted with EtOAc (2X 50 mL), washed with brine (50 mL), na 2 SO 4 Dried, filtered and concentrated in vacuo to give the crude product. The crude product was purified by silica gel column (EtOAc/PE = 1/4) to give impure S-500-2-9 \u2 (560 mg) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.28-5.25(m,1H),3.90-3.80(m,0.25H),3.68-3.58(m,0.75H),2.48-2.36(m,1H),2.05-1.95(m,3H),1.95-1.80(m,1H),1.80-1.75(m,1H),1.75-1.52(m,6H)1.52-1.42(m,6H),1.42-1.32(m,3H),1.32-1.22(m,3H),1.22-1.12(m,3H),1.12-1.02(m,2H),1.01(s,3H),1.00-0.92(m,1H),0.92-0.85(m,9H),0.85-0.77(m,1H),0.69(s,3H)。
S-500-2-9_2 (560 mg) was purified by SFC (column: chiralcel OD-3X 4.6mm I.D.,3um mobile phase: A: CO 2B: ethanol (0.05% DEA) gradient: 5% to 40% B in 5 minutes and held at 40% for 2.5 minutes, then 5% B was held for 2.5 minutes, flow rate: 2.5mL/min, column temperature: 35 ℃) to obtain impure solid 30 (160 mg) and solid 75 (265mg, 47%).
75:
1 H NMR(400MHz,CDCl 3 )δ5.35-5.30(m,1H),3.70-3.60(m,1H),2.50-2.40(m,1H),2.05-1.90(m,4H),1.85-1.75(m,2H),1.75-1.60(m,1H),1.55-1.45(m,8H),1.45-1.25(m,8H),1.25-1.10(m,4H),1.10-1.05(m,2H),1.02(s,3H),0.99-0.91(m,3H),0.91-0.89(m,4H),0.88(s,3H),0.69(s,3H)。
LCMS Rt =1.162 min, chromatography at 1.5 min, 5-95AB, 99% purity, MS ESI C 28 H 45 [M+H-2H 2 O] + Calculated value 381, found value 381 of (1).
Example 76: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((S) -1- (1-hydroxycyclopropyl) ethyl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (76)
Figure BDA0003762336660002321
1. Adding NaClO 2 (374mg, 4.14mmol), TEMPO (645mg, 4.14mmol) and NaClO (10mL, 10% in water) were added to a solution of N-8-7_1 (500mg, 1.38mmol) in MeCN (30 mL). After stirring the mixture at 50 ℃ for 48hrs, a solid appeared. The solid was collected by filtration and triturated with DCM (5 mL) to give N-8-26_1 (180mg, 34%) as a solid.
1 H NMR(400MHz,MeOD)δ2.30-2.20(m,1H),1.98-1.91(m,1H),1.86-1.72(m,1H),1.71-1.63(m,1H),1.62-1.49(m,8H),1.43-1.31(m,4H),1.30-1.20(m,4H),1.19-1.07(m,8H),1.06-0.86(m,8H),0.75-0.65(m,4H)。
2. Will K 2 CO 3 (328mg, 2.38mmol) and MeI (686mg, 4.77mmol) were added to a solution of N-8-26_1 (180mg, 0.477mmol) in DMF (5 mL). After stirring for 16hrs at 20 ℃ the mixture was 50% NH 4 Cl (20 mL) quenched and extracted with EtOAc (3 × 10 mL). The combined organic phases were washed with LiCl (3% in water, 30 mL) and Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-10% etoac in PE) to give N-8-26_2 (160mg, 86%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.63(s,3H),2.45-2.36(m,1H),1.92-1.85(m,1H),1.74-1.58(m,6H),1.56-1.46(m,4H),1.42-1.19(m,9H),1.18-1.15(m,3H),1.13-0.93(m,4H),0.91-0.84(m,4H),0.82(s,3H),0.70-0.61(m,4H)。
3. At 20 ℃ adding Ti (i-PrO) 4 (57.9mg, 0.204mmol) and EtMgBr (0.204mL, 3M in Et 2 O, 0.612 mmol) was added to a solution of N-8-26 \u2 (80mg, 0.204mmol) in THF (2 mL). After stirring for 30 minutes at 20 ℃ the reaction mixture was saturated with NH 4 The Cl (30 mL) solution was quenched and extracted with EtOAc (3 × 20 mL). The combined organic layers were washed with brine (50 mL) and Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain crude productThe material was purified by silica gel column (0-10% EtOAc in PE) to give 76 (1695g, 20%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ1.98-1.86(m,2H),1.69-1.58(m,6H),1.54-1.44(m,3H),1.44-1.29(m,4H),1.28-1.18(m,4H),1.18-1.10(m,5H),1.09-0.93(m,4H),0.91-0.81(m,9H),0.71-0.57(m,6H),0.31-0.24(m,1H)。
LCMS Rt =1.184 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 26 H 41 [M+H] + Calculated value 353 of (1), found value 353.
Example 77: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-ethyl-17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (77)
Figure BDA0003762336660002341
1. A solution of 1-bromo-3-methylbutane (11.7 g, 78mmol) in THF (8 mL) was added dropwise to Mg (4.35g, 179mmol) and I at 60 deg.C 2 (20 mg) suspension in THF (2 mL). The mixture was stirred at 60 ℃ for 1 hour. The mixture was diluted with THF (10 mL) and used directly. In N 2 Freshly prepared isopentylmagnesium bromide (19.5mL, 3.9M in THF, 76 mmol) was added to a solution of S-200-INT-5E (1.0 g, 2.78mmol) in THF (5 mL) at 0 deg.C. The mixture was stirred at 0 ℃ for 1hr. Reacting NH 4 Cl (20 mL, saturated aqueous solution) was added to the mixture. The mixture was extracted with EtOAc (2 × 30 mL). The combined organic phases were washed with brine (100 mL) and Na 2 SO 4 Dried, concentrated in vacuo, purified by silica gel (PE/EtOAc =20/1 to 10/1), and purified from CH 3 CN (10 mL) was recrystallized to give 77 (255mg, 21%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.32-5.26(m,1H),3.66-3.59(m,1H),2.42-2.32(m,1H),2.07-1.85(m,4H),1.77-1.58(m,4H),1.55-1.38(m,10H),1.38-1.19(m,5H),1.19-1.00(m,8H),1.00-0.81(m,13H),0.69(s,3H)。
LCMS Rt =1.306 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 29 H 49 O[M+H-H 2 O] + Calculated value 413, found value 413.
Example 78: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((1S, 2S) -1-cyclopentyl-1-hydroxypropan-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (78)
Figure BDA0003762336660002351
1. Reacting NaBH 4 (550mg, 14.5 mmol) was added to a mixture of N-8-15_2 (240mg, 0.559mmol) in MeOH (3 mL) and THF (2 mL). The mixture was stirred at 15 ℃ for 0.5h. Add another batch of NaBH 4 (550mg, 14.5mmol). The reaction mixture was stirred for an additional 1 hour. Water (5 mL) was added to the reaction mixture. The resulting mixture was extracted with EtOAc (2 × 10 mL). The combined organic layers were washed with brine (10 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-5% EtOAc in PE) to give 58 (7 mg, 5%) and 78 (50 mg, impure), 78 (50 mg, impure) was further purified by flash column (0-5% EtOAc in PE) to give 78 (17mg, 12%) as a solid.
78:
1 H NMR(400MHz,CDCl 3 )δ3.38-3.47(m,1H),2.01-1.82(m,4H),1.71-1.53(m,11H),1.53-1.48(m,4H),1.48-1.30(m,5H),1.30-1.11(m,7H),1.11-0.98(m,5H),0.98-0.85(m,7H),0.85-0.80(m,3H),0.65(s,3H)。
LCMS Rt =1.358 min, chromatography over 2min, 30-90AB _7MIN _E, 100% purity, MS ESI C 29 H 47 [M+H-2H 2 O] + Calculated 395, found 395.
HPLC Rt =6.093 min, at 10 min chromatography, 50-100ab _, 10 min. M, purity 98%.
Example 79: synthesis of (1R, 3S, 4S) -4- ((3S, 5S,8R,9S,10S,13S,14S, 17R) -3-hydroxy-10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-17-yl) -1-phenylpentane-1, 3-diol (79)
Figure BDA0003762336660002361
79 is described in example 13.
79:
1 H NMR(400MHz,CDCl 3 )δ7.43-7.28(m,5H),5.05-4.94(m,1H),4.04-3.91(m,1H),2.51(brs,1H),2.07-1.78(m,6H),1.70-1.61(m,4H),1.51-1.41(m,3H),1.39-1.12(m,11H),1.05-0.98(m,2H),0.91-0.81(m,7H),0.71-0.60(m,4H)。
LCMS Rt =1.298 min, chromatography over 2min, 10-80AB _2MIN _E, purity 96.7%, MS ESI C 31 H 45 F 3 O 3 Na[M+Na] + Calculated value 545, found value 545.
SFC Rt =1.483 min, IC-3 \ u MeOH (DEA) _40 _u2.5ML in 10 min chromatography, 100% de.
13:
1 H NMR(400MHz,CDCl 3 )δ7.40-7.28(m,5H),5.12-5.07(m,1H),3.95-3.88(m,1H),2.76(brs,1H),2.08-1.78(m,6H),1.75-1.60(m,5H),1.51-1.38(m,4H),1.36-1.09(m,9H),1.00-0.89(m,6H),0.83(s,3H),0.71-0.64(m,1H),0.63(s,3H)。
LCMS Rt =1.309 min, chromatography over 2min, 10-80AB _2MIN _E, purity 100%, MS ESI C 31 H 45 F 3 O 3 Na[M+Na] + Calculated 545, found 545.
SFC Rt =1.683 min, IC-3 \ u MeOH (DEA) _40 _u2.5ML, 98.94% de by chromatography over 5 min.
SFC Rt =4.785 min, 8min chromatography, AD _ MEOH (DEA) _5_40_2,8ML _8MIN,94.03% de.
Example 80: synthesis of (3S, 8R,9S,10R,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -3, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (80)
Figure BDA0003762336660002371
1. N-4-14 \u7 (1g, 2.48mmol) was purified by SFC (column: AD (250mm. 30mm, 5um), condition: 0.1% NH 3 H 2 O ETOH, start: b:40%, end B: 40%) to give 80 (Peak 2, 300mg, impure) and 83 (Peak 1, 250mg, impure) as solids. 80 (300 mg, impure) was recrystallized from MeCN (4 mL) at 82 ℃ under reflux for 1 hour. The stirred mixture was cooled to 25 ℃. The suspension was vacuum filtered to give 80 (150mg, 15%) as a solid. 83 (250 mg, impure) was recrystallized from MeCN (3 mL) at 82 ℃ under reflux for 1 h. The stirred mixture was cooled to 25 ℃. The suspension was vacuum filtered to give 83 (150mg, 15%) as a solid.
80:
1 H NMR(400MHz,CDCl 3 )δ5.41-5.39(m,1H),3.63-3.62(m,1H),2.22-1.67(m,10H),1.64-1.36(m,12H),1.16-1.03(m,8H),0.98-0.80(m,11H),0.70(s,3H)。
LCMS Rt =1.276 min, chromatography at 2.0 min, 30-90AB, 99% purity, MS ESI C 27 H 42 [M+H-2H 2 O] + The calculated value 367 and the measured value 367 of (4).
SFC Rt =5.236 min, AD _3_EtOH _DEA5 _40_25ML,100% by 10 min chromatography.
Example 81: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3-Ethyl-10, 13-dimethyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybut-2-yl) -2,3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (81)
Figure BDA0003762336660002381
1. S-500-6-1 \ (350 mg) was purified by SFC (column: AD (250mm. 30mm,5 um), condition: 0.1% NH 3 .H 2 O EtOH, gradient: from 35% to 35%, flow rate (ml/min): 60mL/min,25 ℃) to give 81 (peak 1, 130mg, 37%) and 65 (peak 2, 180mg, 52%) as white solids.
81:
1 H NMR(400MHz,CDCl3)δ5.34-5.24(m,1H),4.09-4.00(m,1H),2.43-2.33(m,1H),2.14(d,J=4Hz,1H),2.07-1.80(m,5H),1.77-1.55(m,5H),1.53-1.30(m,7H),1.28-1.00(m,11H),1.00-0.91(m,1H),0.85(t,J=8Hz,3H),0.70(s,3H)。
LCMS Rt =1.220 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 25 H 38 F 3 O[M+H-H 2 O] + The calculated value 411 of (a), the measured value 411.
SFC _ E1 peak 1: rt =4.561 min, at 10 min chromatography, AD _3_etoh _dea _5_40_25ml ("column: chiralpak AD-3X 4.6mm I.D.,3um mobile phase A: CO 2B: ethanol (0.05% DEA) gradient: 5% to 40% B in 5 minutes and held at 40% for 2.5 minutes, then 5% B for 2.5 minutes, flow rate: 2.5mL/min, column temperature: 35 ℃"), 100% de.
Example 82: synthesis of (3S, 5R,8R,9R,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -13-methylhexahydro-1H-cyclopenta [ a ] phenanthren-3-ol (82)
Figure BDA0003762336660002391
1. Pd/C (anhydrous, 200 mg) was added under Ar to a solution of 44 (200mg, 0.480mmol) in MeOH/THF (10 mL/10 mL). The suspension was degassed under vacuum and washed with H 2 The purge was carried out three times. Mixing the mixture in H 2 Stirring was carried out at 50 ℃ for 48hrs (50 psi) to give a black suspension. The reaction mixture was filtered through a pad of celite and washed with THF (100 mL). The filtrate was concentrated to give solid 28 (30mg, 15%) and solid 82 (30mg, 15%).
82:
1 H NMR(400MHz,CDCl3)δ3.63-3.61(m,1H),2.13-2.00(m,1H),1.99-1.81(m,2H),1.72-1.57(m,6H),1.54-1.34(m,11H),1.33-1.16(m,7H),1.15-0.96(m,5H),0.92-0.85(m,13H),0.81-0.69(m,1H),0.67(s,3H)。
LCMS Rt =1.348 min, chromatography over 2 min, 30-90AB, purity 100%, MS ESI C 28 H 47 [M+H-2H 2 O]+ calculated value 383, found value 383.
Example 83: synthesis of (3S, 8R,9S,10R,13S,14S, 17R) -17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -3, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (83)
Figure BDA0003762336660002401
Figure BDA0003762336660002411
1. N-4-14 \ (u 7) (1g, 2.48mmol) was purified by SFC (column: AD (250mm. 30mm,5 um), condition: 0.1% NH 3 H 2 O ETOH, start: b:40%, end B: 40%) to give 80 (peak 2, 300mg, impure) and 83 (peak 1, 250mg, impure) as solids. 80 (300 mg, impure) was recrystallized from MeCN (4 mL) at 82 ℃ under reflux for 1 hour. The stirred mixture was cooled to 25 ℃. The suspension was vacuum filtered to give 80 (150mg, 15%) as a solid. 83 (250 mg, impure) was recrystallized from MeCN (3 mL) at 82 ℃ under reflux for 1h. The stirred mixture was cooled to 25 ℃. The suspension was vacuum filtered to give 83 (150mg, 15%) as a solid.
83:
1 H NMR(400MHz,CDCl 3 )δ5.41-5.39(m,1H),3.62-3.60(m,1H),2.22-1.89(m,6H),1.64-1.49(m,9H),1.46-1.11(m,16H),0.98-0.86(m,10H),0.70(s,3H)。
LCMS Rt =1.268 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 27 H 42 [M+H-2H 2 O] + The calculated value 367 and the measured value 367 of (4).
SFC Rt =4.609 min, AD _3_EtOH _DEA5 _40 _25MLin 10 min chromatography, 100% b.
Example 84: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -4-cyclopentyl-3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (84)
Figure BDA0003762336660002421
1. Pd (OH) 2 (160 mg, anhydrous) was added to a solution of 32 (80mg, 0.18mmol) in MeOH (20 mL). The mixture was heated at 50 ℃ in H 2 Stirring was carried out for 48 hours under (50 Psi). The mixture was filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 67 (10 mg, 12%) and 84 (30mg, 37%) as solids.
84:
1 H NMR(400MHz,CDCl 3 )δ3.75-3.66(m,1H),2.00-1.90(m,2H),1.86-1.75(m,2H),1.73-1.55(m,11H),1.53-1.26(m,9H),1.25-1.15(m,6H),1.14-1.03(m,5H),1.02-0.92(m,3H),0.91-0.85(m,6H),0.82(s,3H),0.72-0.58(m,4H)。
LCMS Rt =1.518 min, chromatography at 2.0 min, 30-90AB _E, 100% purity, MS ESI C 30 H 49 [M+H-H 2 O] + The calculated value 409 of (1) is actually measured value 409.
Example 85: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (85)
Figure BDA0003762336660002431
1. NaBH is reacted at 15 ℃ 4 (0.96g, 25.4 mmol) was added portionwise to a solution of N-4-1_8 (0.6g, 1.27mmol) in THF (10 mL) and MeOH (5 mL). The mixture was stirred at 15 ℃ for 30mins. Adding NH to the mixture 4 Cl (50mL, 10%). The mixture was extracted with EtOAc (2X 50 mL)) And (4) extracting. The combined organic layers were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo and purified by flash column (0-15% etoac in PE) to give impure 42 and 85. 42 was triturated from MeCN (10 mL) at 15 ℃ and dried in vacuo to give 42 (153mg, 25%) as a solid. 85 was purified by flash column (0-15% EtOAc in PE) to afford an oil, which was treated with MeCN (5 mL) and water (5 mL), which was concentrated in vacuo to afford 85 (70mg, 12%) as a solid.
85:
1 H NMR(400MHz,CDCl 3 )δ3.66-3.55(m,1H),2.10-1.91(m,3H),1.88-1.78(m,1H),1.72-1.55(m,6H),1.50-1.38(m,9H),1.37-0.95(m,10H),0.94-0.79(m,13H),0.75-0.61(m,4H)。
LCMS Rt =1.343 min, chromatography at 2.0 min, 30-90_AB _E, purity 100%, MS ESI C 28 H 46 F 3 O[M+H-H 2 O] + Calculated value 455 of (a), measured value 455.
HPLC Rt =5.14 min, chromatography at 10.0 min, 50-100\uab _e, purity 98.56%.
Example 86: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((1S, 2S) -1-hydroxy-1- (tetrahydro-2H-pyran-4-yl) propan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (86)
Figure BDA0003762336660002441
1. A solution of 4-chlorotetrahydro-2H-pyran (1.2g, 10mmol) in THF (5 mL) was added dropwise to Mg (486mg, 20mmol) and I at 70 deg.C 2 (1 mg) of the mixture. The mixture was stirred at 50 ℃ for 0.5h, diluted with THF (5 mL) and used directly. At 0 ℃ in N 2 Next, a solution of N-8-7 \u1 (500mg, 1.38mmol) in THF (5 mL) was added to a solution of (tetrahydro-2H-pyran-4-yl) magnesium chloride (4.14mL, 1M in THF). The mixture was then stirred at 15 ℃ for a further 18 hours. Reaction mixture with saturated NH 4 Cl (5 mL) was quenched, and the resulting mixture was extracted with EtOAc (2 × 10 mL). The combined organic layers were washed with brine (5 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-15% EtOAc in PE) to give N-8-17_1 (350mg, 57%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.08-3.91(m,2H),3.43-3.31(m,2H),3.31-3.25(m,1H),1.98-1.91(m,2H),1.91-1.80(m,1H),1.70-1.50(m,10H),1.50-1.41(m,2H),1.41-1.32(m,5H),1.32-1.15(m,9H),1.15-0.92(m,6H),0.92-0.83(m,7H),0.65(s,3H).
2. DMP (0.852g, 2.01mmol) was added to a solution of N-8-17 \u1 (300mg, 0.671mmol) in DCM (5 mL). After stirring for 10 min at 15 ℃ the reaction mixture was saturated with NaHCO 3 (20 mL) quench until the pH of the aqueous layer became about 9. The mixture was filtered. The DCM layer was separated and the aqueous phase was extracted with DCM (2 × 20 mL). The combined organic phases were saturated with Na 2 S 2 O 3 Aqueous solution (3 × 20 mL), saturated NaHCO 3 (20 mL), brine (50 mL), washed with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (0-30% etoac in PE) to give N-8-17_3 (200 mg) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.08-3.91(m,2H),3.50-3.31(m,2H),2.73-2.51(m,2H),1.98-1.79(m,1H),1.79-1.42(m,16H),1.42-1.18(m,7H),1.18-0.93(m,8H),0.93-0.79(m,6H),0.68(s,4H)。
3. Subjecting LiAlH to 0 deg.C 4 (50.9mg, 1.34mmol) was added to a mixture of N-8-17-3 (200mg, 0.449mmol) in THF (5 mL). After stirring at 15 ℃ for 0.5h, the reaction mixture was quenched with water (3 mL) and extracted with EtOAc (2X 10 mL). The combined organic layers were washed with brine (10 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-5% EtOAc in PE) to give 86 (23mg, 11%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ4.08-3.91(m,2H),3.41-3.31(m,2H),3.31-3.22(m,1H),2.01-1.79(m,3H),1.70-1.61(m,1H),1.61-1.53(m,8H),1.53-1.51(m,1H),1.51-1.39(m,5H),1.39-1.13(m,8H),1.13-0.92(m,5H),0.92-0.85(m,7H),0.82(s,3H),0.66(s,4H)。
LCMS Rt =4.832 min, chromatography over 7min, 30-90AB 7MIN \ u E, 100% purity, MS ESI C 29 H 47 O[M+H-2H 2 O] + The calculated value 411 of (a), the actual measurement value 411.
HPLC Rt =6.338 minutes, chromatography over 10 minutes, 30-90ab _1.2ml e.met,100% purity.
Example 87: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3R) -4- (4, 4-dimethylcyclohexyl) -3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (87)
Figure BDA0003762336660002461
1. Pd (OH) 2 (150 mg, anhydrous) was added to a solution of 12 (100mg, 0.206mmol) in MeOH (20 mL). The mixture was heated at 50 ℃ in H 2 Next (50 Psi) was stirred for 48 hours. The mixture was filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 87 (12mg, 12%) and 20 (11mg, 11%) as solids.
87:
1 H NMR(400MHz,CDCl 3 )δ3.82-3.75(m,1H),2.00-1.83(m,2H),1.80-1.58(m,7H),1.52-1.42(m,4H),1.40-1.27(m,10H),1.25-1.14(m,10H),1.13-0.98(m,6H),0.96(s,3H),0.94-0.82(m,12H),0.67(s,3H)。
LCMS Rt =1.734 min, chromatography 2.0 min, 30-90AB _E, 100% purity, MS ESI C 33 H 55 [M+H-2H 2 O] + Calculated value of (451), found value of 451.
Example 88: synthesis of (3S, 5S,8R,9R,10S,13S,14S, 17R) -17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -3, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (88)
Figure BDA0003762336660002471
1. At Ar lower Pd (OH) 2 (200 mg) was added to a solution of 83 (150mg, 0.372mmol) in MeOH (3 mL) and THF (3 mL). The suspension is degassed in vacuo and treated with H 2 The purge was carried out three times. Mixing the mixture in H 2 (50 psi) stirred at 50 ℃ for 12h to give a black suspension. The reaction mixture was filtered through a pad of celite and washed with DCM (3 × 50 mL). The filtrate was concentrated in vacuo to provide an oil. The residue was purified by silica gel chromatography (PE/EtOAc =8/1 to 5/1) to give 88 (18mg, 12%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.62-3.59(m,1H),1.97-1.81(m,2H),1.76-1.50(m,12H),1.46-1.28(m,5H),1.24-1.03(m,12H),0.95-0.83(m,11H),0.74-0.57(m,5H)。
LCMS Rt =1.317 min, chromatography at 2.0 min, 30-90AB, 99% purity, MS ESI C 27 H 44 [M+H-2H 2 O] + Found 369, found 369.
Example 89: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((1S, 2S) -1-hydroxy-1- (pyridin-3-yl) propan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (89)
Figure BDA0003762336660002472
Figure BDA0003762336660002481
1.N-8-19- \ u 1 (3340) (100mg, 0.227mmol) was separated by SFC (column: AD (250mm. 30mm, 5um), gradient: 50-50% B (A =0.05% NH) 3 /H 2 O, B = MeOH), flow rate: 80 mL/min) to give 7 (Peak 1, 57mg, 57%) and 89 (Peak 2,8mg, 8%) as solids.
SFC peak 1: rt =1.798 min and peak 2Rt =1.985 min, 3 min chromatography, AD-H _3UM _4_5_40_4ML ("Chiralpak AD-3 50 x 4.6mm I.D.,3um mobile phase: A: CO 2 B: isopropanol (0.05% DEA) gradient: 5% to 40% B at 1.4 minutes and at 40% for 1.05 minutes, then 5% b retention 0.35 min, flow rate: 4mL/min, column temperature: 40 ℃ C. ").
Example 90: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((1R, 2S) -1-cyclopropyl-1-hydroxypropan-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (90)
Figure BDA0003762336660002482
Figure BDA0003762336660002491
1. Reacting NaBH 4 (1.18g, 17.4mmol) was added five times, every 5 minutes, to a solution of N-8-13_2 (140mg, 0.347mmol) in MeOH (1 mL) and THF (1 mL). The mixture was stirred at 15 ℃ for 30 minutes. Mixing the mixture with sat 4 Cl (50 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases are washed with Na 2 SO 4 Dried, filtered, concentrated and purified by combi-flash (25% etoac in PE) to give solid 10 (26mg, 19%) and solid 90 (12mg, 9%).
90:
1 H NMR(400MHz,CDCl 3 )δ3.00-2.80(m,1H),2.01-1.95(m,1H),1.75-1.60(m,5H),1.47-1.18(m,13H),1.15-0.79(m,18H),0.70-0.60(m,4H),0.58-0.50(m,1H),0.48-0.40(m,1H),0.38-0.30(m,1H),0.24-0.16(m,1H)。
LCMS Rt =3.796 min, 7.0 min chromatography, 30-90AB 7MIN \ E, 100% purity, MS ESI C 27 H 43 [M+H-2H 2 O] + The calculated value 367 and the actual value 367 of (a).
HPLC Rt =13.689 min, chromatography over 30min, 70-90ab \u1/min. M, 98% pure.
Example 91: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxybut-2-yl) -10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (91)
Figure BDA0003762336660002492
The synthesis of 91 is described in example 13.
91:
1 H NMR(400MHz,CDCl 3 )δ3.98-3.88(m,1H),2.11-2.02(m,1H),2.00(s,1H),1.98-1.88(m,2H),1.85-1.79(m,1H),1.73-1.58(m,4H),1.52-1.20(m,11H),1.19-1.11(m,4H),1.10-1.00(m,3H),0.97-0.89(m,4H),0.85(s,3H),0.75-0.68(m,1H),0.66(s,3H)。
LCMS Rt =1.155 min, chromatography at 2.0 min, 30-90_AB _E, 100% purity, MS ESI C 24 H 38 F 3 O[M+H-H 2 O] + Calculated value 399, actually measured value 399.
HPLC Rt =5.23 min, chromatography at 10.0 min, 30-90 \uab _e, purity 98.88%, d.e.100%.
Example 92: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -3-hydroxy-6-methylhept-2-yl) -3,10, 13-trimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (92)
Figure BDA0003762336660002501
1. DMP (2.42g, 5.72mmol) was added to a solution of N-4-16/1 (1.00g, 2.86mmol) in DCM (20 mL). The reaction was then stirred at 15 ℃ for 10 minutes. Saturated NaHCO 3 Aqueous solution (20 mL) solution and saturated Na 2 S 2 O 3 Aqueous (20 mL) solution was added to the reaction mixture, which was then extracted with DCM (2 × 20 mL). The combined organic layers were washed with saturated NaHCO 3 Aqueous solution (3X 60 mL) and brine (60 mL) were washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo to give a solid. The residue was purified by silica gel chromatography (PE/EtOAc =0 to 30%) to give N-4-16 \ u 2 (800mg, 81%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ9.58-9.53(m,1H),2.41-2.31(m,1H),1.96-1.81(m,4H),1.89-1.34(m,10H),1.32-1.21(m,10H),1.16-1.09(m,5H),0.97(s,3H),0.89-0.84(m,1H),0.69(s,3H)。
2. At 25 ℃ in N 2 A solution of N-4-16 (300mg, 0.86mmol) in THF (10 mL) was added to a solution of isoamyl magnesium bromide (4.32mL, 2M in ether, 8.65 mmol). The mixture was stirred at 25 ℃ for 30 minutes and passed through saturated NH 4 Cl (10 mL) quenched and extracted with ethyl acetate (3 × 10 mL). The organic layer was washed with brine (20 mL) and Na 2 SO 4 Dried and filtered, concentrated in vacuo and purified by flash column (0-15% EtOAc in PE) to give N-4-16 (200 mg, impure) as a solid, which was triturated in MeCN (10 mL) at 25 ℃ to give 92 (141mg, 70%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ3.67-3.57(m,1H),2.01-1.77(m,4H),1.67-1.57(m,4H),1.55-1.26(m,14H),1.25-1.21(m,5H),1.19-0.99(m,7H),0.96(s,3H),0.93-0.84(m,9H),0.66(s,3H)。
LCMS Rt =1.367 min, chromatography for 2min, 30-90AB _2MIN _E, purity 100%, MS ESI C 28 H 47 [M+H-2H 2 O] + Calculated value 383 of (d), found value 383.
Example 93: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-4-phenylbutan-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (93)
Figure BDA0003762336660002511
Figure BDA0003762336660002521
1. A mixture of N-6-5 and N-6-6 (190mg, 0.420mmol) was separated by prep.HPLC (column: YMC-Actus Triart C18 x 30mm x 5um; conditions: water (0.05% HCl) -ACN; gradient: 90-100% B; flow rate: 25 mL/min) to give 93 (56mg, 30%) as a solid and 32 (12mg, 6%) as a solid.
93:
1 H NMR(400MHz,CDCl 3 )δ7.34-7.28(m,2H),7.25-7.18(m,3H),3.95-3.86(m,1H),2.87-2.75(m,1H),2.69-2.58(m,1H),2.01-1.91(m,1H),1.90-1.79(m,1H),1.69-1.58(m,4H),1.55-1.41(m,6H),1.40-1.11(m,11H),1.07-0.95(m,6H),0.91-0.80(m,7H),0.69-0.59(m,4H)。
LCMS Rt =1.334 min, chromatography at 2.0 min, 30-90AB, 100% purity, MS ESI C 31 H 48 O 2 Na[M+Na] + Calculated value 475, found value 475.
Example 94: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -4-cyclopentyl-3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (94)
Figure BDA0003762336660002522
Figure BDA0003762336660002531
1. Pd (OH) 2 (300 mg, anhydrous) was added to a solution of 100 (150mg, 0.338mmol) in MeOH (20 mL). The mixture was heated at 50 ℃ in H 2 Stirring was continued for 48 hours at 50 psi. The mixture was filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 94 (6 mg, 4%) and 98 (46mg, 30%) as solids.
94:
1 H NMR(400MHz,CDCl 3 )δ3.78-3.66(m,1H),1.98-1.72(m,7H),1.69-1.59(m,4H),1.48-1.32(m,12H),1.27-1.07(m,12H),1.06-1.00(m,3H),0.97(s,3H),0.94-0.85(m,7H),0.66(s,3H)。
LCMS Rt =1.639 min, chromatography 2.0 min, 30-90AB _E, 98.8% purity, MS ESI C 30 H 49 [M+H-2H 2 O] + The calculated value 409 of (1) is actually measured value 409.
Example 95: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -3- (methoxymethyl) -10, 13-dimethyl-17- ((2S, 3R) -4, 4-trifluoro-3-hydroxybut-2-yl) -2,3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (95)
Figure BDA0003762336660002532
Figure BDA0003762336660002541
1.N-004-029A (0.45g, 1.01mmol) was purified by SFC (column: AD (250mm. Multidot.30mm, 5 um), condition: 0.1% NH 3 H 2 O ETOH, start B:30%, end B: 30%) to give white solid 39 (PK 1:120mg, 26.7%) and white solid 95 (PK 2:200mg, 44.5%).
95:
1 H NMR(400MHz,CDCl 3 )δ5.32-5.29(m,1H),4.06-3.99(m,1H),3.37(s,3H),3.30-3.19(m,2H),2.54(s,1H),2.42-2.33(m,1H),2.17-2.07(m,1H),2.20-1.85(m,5H),1.77-1.63(m,4H),1.51-0.83(m,17H),0.73(s,3H)。
LCMS Rt =1.103 min, chromatography over 2min, 30-90AB _2MIN _E, 100% purity, MS ESI C 24 H 34 F 3 O[M-CH 5 O 2 ] + Calculated 395, found 395.
Example 96: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3-Ethyl-17- ((2S, 3R) -3-hydroxy-6-methylhept-2-yl) -10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (96)
Figure BDA0003762336660002542
Figure BDA0003762336660002551
1. Pd (OH) 2 (200 mg) was added to a solution of 52 (50mg, 0.116mmol) in MeOH (10 mL). The mixture was heated at 50 ℃ in H 2 Stirring was performed under (50 Psi). The mixture was filtered, concentrated and purified by combi-flash (0-10% EtOAc in PE) to give 24 (15mg, 30%) and 96 (1.2mg, 3%) solids.
96:
1 H NMR(400MHz,CDCl 3 )δ3.67-3.55(m,1H),2.01-1.83(m,2H),1.80-1.62(m,4H),1.61-1.56(m,2H),1.55-1.50(m,1H),1.49-1.31(m,10H),1.30-1.10(m,11H),1.09-1.00(m,3H),0.96(s,3H),0.94-0.86(m,12H),0.67(s,3H)。
LCMS t R =1.326 min, chromatography over 2 min, 30-90ab _elsd, purity 100.0%, MS ESI C 29 H 49 [M+H-2H 2 O] + Calculated value 397 of (a), found value 397.
Example 97: synthesis of (3S, 5R,8R,9S,10S,13S,14S, 17R) -3,10, 13-trimethyl-17- ((2S, 3S) -4, 4-trifluoro-3-hydroxybutan-2-yl) hexahydro-1H-cyclopenta [ a ] phenanthren-3-ol (97)
Figure BDA0003762336660002552
1. To a solution of N-004-017 (100mg, 0.19mmol) in THF (2 mL) and MeOH (1 mL) and water (1 mL) was added KOH (53.8mg, 0.96mmol). The mixture was stirred at 60 ℃ for 16 hours. The mixture was poured into water (20 mL) and extracted with EtOAc (2 × 40 mL). The combined organic layers were washed with brine (30 mL) and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by flash column (0-15% etoac in PE) to give 97 (80 mg, impure) as a white solid. 97 (80mg, 0.19mmol) was purified through flash column (0-10% EtOAc in PE) to give 97 (60 mg, impure). At 0 ℃ in N 2 NaBH was added in one portion to a solution of 97 (40mg, 0.096 mmol) in methanol (3 mL) 4 (10.8mg, 0.28mmol). The mixture was stirred at 0 ℃ for 30 minutes. The mixture was poured into water (10 mL) and stirred for 20 min. The aqueous phase was extracted with EtOAc (3 × 10 mL). The combined organic phases were washed with brine (2X 20 mL)) Washing with Na 2 SO 4 Dried, filtered and concentrated to give the crude product, which was combined with another batch derived from 20mg of impure 97, and the residue was purified by flash column (0-10% etoac in PE) to give 97 (31mg, 31%) as a white solid.
1 H NMR(400MHz,CDCl3)δ4.11-3.96(m,1H),2.18-2.11(d,J ab =6.4Hz,1H),2.02-1.77(m,5H),1.68-1.57(m,3H),1.49-1.24(m,11H),1.23-1.19(m,5H),1.18-1.01(m,7H),0.96(s,3H),0.67(s,3H)。
LCMS Rt =1.124 min, chromatography over 2min, 30-90AB _2MIN _E.M, 100% purity, MS ESI C 24 H 38 F 3 O[M+H-H 2 O] + Calculated value 399, actually measured value 399.
Example 98: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S) -4-cyclopentyl-3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (98)
Figure BDA0003762336660002561
Figure BDA0003762336660002571
1. Pd (OH) 2 (300 mg, anhydrous) was added to a solution of 100 (150mg, 0.338mmol) in MeOH (20 mL). The mixture was heated at 50 ℃ in H 2 Stirring was continued for 48 hours at 50 psi. The mixture was filtered, concentrated and purified by combi-flash (0-15% etoac in PE) to give 94 (6 mg, 4%) and 98 (46mg, 30%) as solids.
98:
1 H NMR(400MHz,CDCl 3 )δ3.78-3.67(m,1H),1.97-1.74(m,6H),1.68-1.56(m,8H),1.53-1.45(m,4H),1.44-1.31(m,10H),1.28-1.21(m,1H),1.16-0.96(m,9H),0.91-0.85(m,6H),0.82(s,3H),0.69-0.61(m,4H)。
LCMS Rt =1.582 min to2.0 min chromatography, 30-90AB _E, 100% purity, MS ESI C 30 H 49 [M+H-2H 2 O] + The calculated value 409 of (1) is actually measured value 409.
Example 99: synthesis of (3S, 5S,8R,9S,10S,13S,14S, 17R) -17- ((2S, 3S, E) -3-hydroxy-5-phenylpent-4-en-2-yl) -10, 13-dimethyl-3- (trifluoromethyl) hexadecahydro-1H-cyclopenta [ a ] phenanthren-3-ol (99)
Figure BDA0003762336660002572
Figure BDA0003762336660002581
1. NaBH is reacted at 20 ℃ 4 (419mg, 11.1mmol) was added portionwise to a solution of N-003-005_1 (140mg, 0.278mmol) in THF (2 mL) and MeOH (1 mL). The mixture was stirred at 20 ℃ for 10 minutes. The reaction was performed using water (20 mL) and NH 4 Cl (20mL, sat.). The mixture was extracted with EtOAc (50 mL). The organic layer was concentrated in vacuo and purified by prep-TLC (PE/EtOAc = 4/1) to give N-003-005 (50 mg, impure) and 14 (50 mg), both as solids.
Impure 99 (50 mg) was purified by SFC (instrument: SFC 1; column: OD (250mm. About. 30mm,5 um); conditions: 0.1% NH 3 H 2 O EtOH; and starting B:40 percent; and B, ending: 40 percent; flow rate (mL/min): 50; injecting: 60 To provide a solid, which was dissolved in MeCN (20 mL) and concentrated in vacuo to give 99 (17 mg) as a solid.
99:
1 H NMR(400MHz,CDCl 3 )δ7.44-7.36(m,2H),7.36-7.28(m,2H),7.25-7.20(m,1H),6.58(d,J=16.0Hz,1H),6.24(dd,J=4.8,16.0Hz,1H),4.49-4.40(m,1H),2.09-1.91(m,4H),1.86-1.75(m,1H),1.72-1.58(m,5H),1.52-1.04(m,14H),0.94(d,J=6.4Hz,3H),0.91-0.87(m,1H),0.86(s,3H),0.75-0.70(m,1H),0.69(s,3H)。
LCMS Rt =1.280 min, chromatography over 2min, 30-90AB _2MIN _E, purity98.5%,MS ESI C 31 H 42 F 3 O[M+H-H 2 O] + The calculated value 487 of (a), the actual measured value 487.
HPLC Rt =6.29 min, chromatography over 8 min, 30-90 μ ab \u1.2 ml,100% d.e.
Example 100: synthesis of (3S, 8S,9S,10R,13S,14S, 17R) -17- ((2S, 3S) -4-cyclopentyl-3-hydroxybut-2-yl) -3-ethyl-10, 13-dimethyl-2, 3,4,7,8,9,10,11,12,13,14,15,16, 17-decatetrahydro-1H-cyclopenta [ a ] phenanthren-3-ol (100)
Figure BDA0003762336660002591
1. A solution of (bromomethyl) cyclopentane (1.8g, 11.0mmol) in THF (11 mL) was added dropwise to Mg (528mg, 22.0mmol) and I at 75 deg.C 2 (55.8mg, 0.22mmol) in THF (3 mL). The mixture was stirred at 75 ℃ for 1 hour. A mixture of (cyclopentylmethyl) magnesium bromide (11.1mL, 11.1mmol,1M in THF) was slowly added to a solution of S-500-6-1 \ (800mg, 2.23mmol) in THF (30 mL) at 15 ℃. After the addition, the mixture was stirred at 15 ℃ for 1 hour. The mixture was washed with saturated NH 4 Cl (40 mL) quenched and extracted with EtOAc (3 × 20 mL). The combined organic phases were washed with brine (2X 30 mL) and Na 2 SO 4 Dried, filtered and concentrated and purified by combi-flash (0-15% etoac in PE) to give 100 (350mg, 35%) as a solid.
1 H NMR(400MHz,CDCl 3 )δ5.32-5.26(m,1H),3.77-3.69(m,1H),2.41-2.31(m,1H),2.09-1.89(m,4H),1.88-1.69(m,4H),1.68-1.55(m,6H),1.54-1.27(m,12H),1.26-1.15(m,2H),1.14-1.05(m,4H),1.04-0.99(m,5H),0.98-0.88(m,4H),0.87-0.81(m,3H),0.69(s,3H)。
LCMS Rt =5.661 min, chromatography over 7.0 min, 30-90AB _E, 100% purity, MS ESI C 30 H 47 [M+H-2H 2 O] + Calculated value 407 of (d), observed value 407.
Table 1. Data for exemplary compounds.
Figure BDA0003762336660002601
Figure BDA0003762336660002611
Figure BDA0003762336660002621
Figure BDA0003762336660002631
Figure BDA0003762336660002641
Figure BDA0003762336660002651
Figure BDA0003762336660002661
Figure BDA0003762336660002671
Figure BDA0003762336660002681
Figure BDA0003762336660002691
Figure BDA0003762336660002701
Figure BDA0003762336660002711
For Table 1, "A" represents EC 50 Is 1 to 100nM, "B" denotes EC 50 Greater than 100nM up to 1. Mu.M, "C" means EC 50 Greater than 1 μ M; "D" represents E Max At most 100%, "E" means E max Between 100% and 500%, "F" denotes E max Greater than 500%; "G" means between 10% and-10% and includes 10% and-10%% of the% enhancement, "H" means less than-10% and greater than or equal to-40% of the% enhancement, and "I" means less than-40% of the% enhancement.
Other embodiments
In the claim elements, such as "a", "an" and "the" may mean one or more than one unless the context clearly dictates otherwise. Claims or descriptions that include "or" between one or more members of a group are deemed to be satisfied if one, more than one, or all of the members of the group are present in, used in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of a group is present in, used in, or associated with a given product or process. The invention includes embodiments in which more than one member of a group, or all members of a group, are present in, used in, or associated with a given product or process.
Furthermore, the present invention encompasses all variations, combinations, and permutations in which one or more limitations, elements, clauses, and descriptive terms from one or more of the listed claims are introduced into another claim. For example, any claim that is independent of other claims may be amended to include one or more limitations in any other claim that refers to the same base claim. When an element is represented according to the listed form, such as in markush, each subgroup of the element is also disclosed, and any element can be removed from the group. It will be understood that, in general, when the invention or aspects of the invention are referred to as comprising particular elements and/or features, some embodiments of the invention or aspects of the invention consist of, or consist essentially of, those elements and/or features. For the purpose of brevity, these embodiments have not been described in detail herein with the same language. It should also be noted that the terms "comprising" and "containing" are intended to be open-ended and allow for the inclusion of other elements or steps. When ranges are given, endpoints are included. Furthermore, unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or sub-range within the stated range, to the tenth of the unit of the value at the lower limit of the range, in different embodiments of the invention, unless the context clearly dictates otherwise.
Various issued patents, published patent applications, journal articles and other publications are referred to herein, all of which are incorporated herein by reference. In case of conflict between a cited document and the present specification, including all references cited herein, shall control. Furthermore, any particular embodiment of the invention that falls within the prior art may be explicitly excluded from any one or more of the claims. As this embodiment is considered known to those skilled in the art, they may be excluded even if not explicitly indicated to be excluded herein. Any particular embodiment of the invention may be excluded from any claim for any reason, whether or not related to the presence of prior art.
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many variations of the specific embodiments described herein. The scope of the embodiments described herein is not intended to be limited by the foregoing description, but rather should be controlled by the appended claims. It will be understood by those skilled in the art that various changes and modifications may be made in the specification without departing from the spirit or scope of the invention as defined in the claims.
The invention also relates to:
1. a compound of formula (I-63):
Figure BDA0003762336660002721
or a pharmaceutically acceptable salt thereof, wherein:
R 1 is an alkyl group;
R 2 is alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl or heteroaryl and R 3 Is hydrogen, alkyl, carbocyclyl, heterocyclyl, aryl or heteroaryl, or
R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring;
R 4 and R 5 Each independently is hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl, or
R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group;
R 6 absent or hydrogen; and is
Figure BDA0003762336660002731
Represents a single or double bond, wherein
Figure BDA0003762336660002732
When one is a double bond, the other is
Figure BDA0003762336660002733
Is a single bond and R 6 Is absent; when two are provided
Figure BDA0003762336660002734
When all are single bonds, then R 6 Is hydrogen.
2. A compound of embodiment 1 wherein R 1 Is an alkyl group.
3. A compound of embodiment 1 wherein R 1 Is an unsubstituted alkyl group.
4. A compound of embodiment 1 wherein R 1 Is C 1 -C 6 An alkyl group.
5. A compound of embodiment 1 wherein R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 An alkyl group.
6. A compound of embodiment 1 wherein R 1 is-CH 3 、–CF 3 or-CH 2 CH 3 or-CH 2 OCH 3
7. A compound of embodiment 1 wherein R 2 Is hydrogen or alkyl.
8. A compound of embodiment 1 wherein R 2 Is unsubstituted C 1 -C 6 Alkyl or C 1 -C 6 A haloalkyl group.
9. A compound of embodiment 1 wherein R 2 Is an unsubstituted alkyl group.
10. A compound of embodiment 1 wherein R 2 Is carbocyclylalkyl.
11. A compound of embodiment 1 wherein R 2 Is an aralkyl group.
12. A compound of embodiment 1 wherein R 2 Is a heterocyclylalkyl group.
13. A compound of embodiment 1 wherein R 2 Is unsubstituted C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, carbocyclyl, carbocyclylalkyl, aralkyl, or heterocyclylalkyl.
14. A compound of embodiment 1 wherein R 2 Is C 1 -C 6 A haloalkyl group.
15. A compound of embodiment 1 wherein R 2 Is unsubstituted C 1 -C 6 An alkyl group.
16. A compound of embodiment 1 wherein R 2 Is a pyridyl group.
17. A compound of embodiment 1 wherein each R 2 Is isopentyl and R 3 Is hydrogen.
18. A compound of embodiment 1 wherein R 2 Is C 5 Alkyl or and R 3 Is hydrogen.
19. A compound of embodiment 1 wherein each R 2 Is isopentyl and R 3 Is hydrogen.
20. A compound of embodiment 1 wherein R 2 is-CF 3 or-CH 3 And R is 3 Is hydrogen.
21. A compound of embodiment 1 wherein R 2 is-CF 3 And R is 3 Is hydrogen.
22. A compound of embodiment 1 wherein R 4 is-OH or halo.
23. A compound of embodiment 1 wherein R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group.
24. A compound of embodiment 1 wherein R 4 Is hydrogen and R 5 Is halo.
25. A compound of embodiment 1 wherein R 4 And R 5 Is halo.
26. A compound of embodiment 1 wherein R 4 And R 5 Is hydrogen.
27. A compound of embodiment 1 wherein R 2 Is aryl or heteroaryl and R 3 Is hydrogen.
28. A compound of embodiment 1 wherein R 2 Is carbocyclyl or heterocyclyl and R 3 Is hydrogen.
29. A compound of embodiment 1 wherein R 2 Is isoamyl and R 3 Is hydrogen.
30. A compound of embodiment 1 wherein R 2 is-CF 3 or-CH 3 And R is 3 Is hydrogen or-CH 3
31. A compound of embodiment 1 wherein R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered carbocyclic or heterocyclic ring.
32. A compound of embodiment 1 wherein R 1 is-CH 3 or-CH 2 CH 3 ,R 2 Is isopentyl, and R 3 Is hydrogen.
33. A compound of embodiment 1 wherein R 1 is-CH 3 or-CH 2 CH 3 ,R 2 Is unsubstituted isopentyl, and R 3 Is hydrogen.
34. The compound of embodiment 1 wherein the compound of formula (I-63) is selected from compounds of formula (I-A63), (I-B63), or (I-C63):
Figure BDA0003762336660002741
35. The compound of embodiment 1, wherein the compound of formula (I-63) is selected from compounds of formula (I-A63):
Figure BDA0003762336660002751
36. the compound of embodiment 1, wherein the compound of formula (I-63) is selected from compounds of formula (I-C63):
Figure BDA0003762336660002752
37. the compound of embodiment 1, wherein the compound of formula (I-63) is selected from compounds of formula (I-D63):
Figure BDA0003762336660002753
38. the compound of embodiment 1, wherein the compound of formula (I-63) is selected from compounds of formula (I-E63):
Figure BDA0003762336660002754
39. the compound of embodiment 1, wherein the compound of formula (I-63) is selected from compounds of formula (I-D-I63) or (I-D-ii 63):
Figure BDA0003762336660002755
Figure BDA0003762336660002761
40. the compound of embodiment 1 wherein the compound of formula (I-63) is selected from compounds of formula (I-E-I63) or (I-E-ii 63):
Figure BDA0003762336660002762
41. a compound of formula (I-67):
Figure BDA0003762336660002763
or a pharmaceutically acceptable salt thereof, wherein:
R 1 is an alkyl group;
R 2 and R 3 Each independently hydrogen, alkyl, carbocyclyl, heterocyclyl, aryl or heteroaryl, or
R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring;
R 4 and R 5 Each independently hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl, or
R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group;
R 6 absent or hydrogen; and is
Figure BDA0003762336660002764
Represents a single or double bond, wherein
Figure BDA0003762336660002765
When one is a double bond, the other is
Figure BDA0003762336660002766
Is a single bond and R 6 Is absent; and when two are
Figure BDA0003762336660002771
When all are single bonds, then R 6 Is hydrogen.
42. A compound of embodiment 41 wherein R 1 Is an unsubstituted alkyl group.
43. A compound of embodiment 41 wherein R 1 Is an unsubstituted alkyl group.
44. A compound of embodiment 41 wherein R 1 Is C 1 -C 6 An alkyl group.
45. A compound of embodiment 41 wherein R 1 is-CH 3 、–CF 3 or-CH 2 CH 3
46. A compound of embodiment 36 wherein R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 An alkyl group.
47. A compound of embodiment 36 wherein R 2 Is hydrogen or alkyl.
48. A compound of embodiment 36 wherein R 2 And R 3 Each independently hydrogen or alkyl.
49. A compound of embodiment 1 wherein R 2 And R 3 Each independently is hydrogen or C 1 -C 6 A haloalkyl group.
50. A compound of embodiment 36 wherein R 2 And R 3 Each independently is hydrogen, -CF 3 or-CH 3
51. A compound of embodiment 36 wherein R 4 is-OH or halo.
52. A compound of embodiment 36 wherein R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group.
53. A compound of embodiment 36 wherein R 4 Is hydrogen and R 5 Is halo.
54. A compound of embodiment 36 wherein R 4 And R 5 Is halo.
55. A compound of embodiment 36 wherein R 4 And R 5 Is hydrogen.
56. A compound of embodiment 36 wherein R 2 Is aryl or heteroaryl and R 3 Is hydrogen.
57. A compound of embodiment 36 wherein R 2 Is carbocyclyl or heterocyclyl and R 3 Is hydrogen.
58. A compound of embodiment 36 wherein R 2 And R 3 Is hydrogen.
59. A compound of embodiment 36 wherein R 2 And R 3 Together with the carbon atoms to which they are attached form a 3-8 membered carbocyclic or heterocyclic ring.
60. The compound of embodiment 36, wherein the compound of formula (I-67) is selected from compounds of formula (I-A67), (I-B67), or (I-C67):
Figure BDA0003762336660002772
Figure BDA0003762336660002781
61. the compound of embodiment 36, wherein the compound of formula (I-67) is selected from compounds of formula (I-A67):
Figure BDA0003762336660002782
62. the compound of embodiment 36 wherein the compound of formula (I-67) is selected from compounds of formula (I-C67):
Figure BDA0003762336660002783
63. a compound selected from:
Figure BDA0003762336660002784
Figure BDA0003762336660002791
Figure BDA0003762336660002801
Figure BDA0003762336660002811
Figure BDA0003762336660002821
Figure BDA0003762336660002831
Figure BDA0003762336660002841
Figure BDA0003762336660002851
Figure BDA0003762336660002861
Figure BDA0003762336660002871
Figure BDA0003762336660002881
64. a pharmaceutical composition comprising a compound of any one of embodiments 1 to 63, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
65. A method of treating or preventing a disease described herein, comprising administering to a subject in need thereof an effective amount of a compound of any one of embodiments 1 to 63, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of embodiment 64.
66. The method according to embodiment 65, wherein said disease is a Gastrointestinal (GI) disorder, a structural disorder affecting the GI tract, an anal disorder, colonic polyps, cancer or colitis.
67. The method according to embodiment 65, wherein the disease is inflammatory bowel disease.
68. The method according to embodiment 65, wherein the disease is cancer, diabetes or sterol synthesis disorders.
69. The method according to embodiment 65, wherein the disease is a metabolic disease.
70. The method according to embodiment 65, wherein the disease is an autoimmune disease.
71. The method according to embodiment 65, wherein the disease is rheumatoid arthritis, juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis, crohn's disease, ulcerative colitis, and plaque psoriasis.
72. A method of treating or preventing a CNS-related disorder, comprising administering to a subject in need thereof an effective amount of a compound of any one of embodiments 1 to 57, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
73. A method according to embodiment 72, wherein said CNS-related disorder is an adjustment disorder, anxiety disorder (including obsessive-compulsive disorder, post-traumatic stress disorder, and social phobia), cognitive disorder, separation disorder, eating disorder, mood disorder, schizophrenia or other psychotic disorder, sleep disorder, substance-related disorder, personality disorder, autism spectrum disorder, neurodevelopmental disorder, multiple sclerosis, sterol synthesis disorder, pain, encephalopathy secondary to a medical condition, epilepsy, stroke, traumatic brain injury, movement disorder, vision disorder, hearing loss, or tinnitus.
74. The method according to embodiment 72, wherein the disease is a disorder of sterol synthesis.
75. A method of effecting negative allosteric modulation of NMDA receptors in a subject in need thereof, comprising administering to the subject a compound of formula (a):
Figure BDA0003762336660002891
or a pharmaceutically acceptable salt thereof, wherein:
R 1 is hydrogen or alkyl;
R 2 and R 3 Each independently is hydrogen, alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl or heteroaryl, or R 2 And R 3 Together with the carbon atom to which they are attached form a 3-8 membered ring;
R 4 and R 5 Each independently is hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or C 1 -C 6 Alkyl, or R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group;
R 6 absent or hydrogen;
R G is hydrogen or alkyl; and is
Figure BDA0003762336660002892
Represents a single or double bond, wherein
Figure BDA0003762336660002893
When one is a double bond, the other is
Figure BDA0003762336660002894
Is a single bond and R 6 Is absent; and whenTwo are provided
Figure BDA0003762336660002895
When all are single bonds, then R 6 Is hydrogen.
76. The method of embodiment 75, wherein R 1 Is an alkyl group.
77. The method of embodiment 75, wherein R 1 Is C 1 -C 6 An alkyl group.
78. The method of embodiment 75 wherein R 1 is-CH 3 、–CF 3 or-CH 2 CH 3
79. The method of embodiment 75, wherein R 1 is-CH 2 OR A Wherein R is A Is C 1 -C 6 An alkyl group.
80. The method of embodiment 75, wherein R 2 Is hydrogen or alkyl.
81. The method of embodiment 75, wherein R 2 And R 3 Each independently hydrogen or alkyl.
82. The method of embodiment 75, wherein R 2 And R 3 Each independently is hydrogen or C 1 -C 6 A haloalkyl group.
83. The method of embodiment 75 wherein R 2 And R 3 Each independently is hydrogen, -CF 3 or-CH 3
84. The method of embodiment 75, wherein R 4 is-OH or halo.
85. The method of embodiment 75 wherein R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group.
86. The method of embodiment 75 wherein R 4 Is hydrogen and R 5 Is halo.
87. The method of embodiment 75, wherein R 4 And R 5 Is halo.
88. The method of embodiment 75, wherein R 4 And R 5 Is hydrogen.
89. The method of embodiment 75Wherein R is 2 Is aryl or heteroaryl and R 3 Is hydrogen.
90. The method of embodiment 75, wherein R 2 Is carbocyclyl or heterocyclyl and R 3 Is hydrogen.
91. The method of embodiment 75, wherein R 2 And R 3 Is hydrogen.
92. The method of embodiment 75, wherein R G Is hydrogen or-CH 3
93. The method of embodiment 75, wherein R 6 Is hydrogen and
Figure BDA0003762336660002901
is a single bond.
R 1 Is an alkyl group.

Claims (10)

1. A compound of formula (I-63):
Figure FDA0003762336650000011
or a pharmaceutically acceptable salt thereof, wherein:
R 1 Is substituted or unsubstituted alkyl;
R 2 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;
R 3 is a substituted or unsubstituted alkyl group, a substituted or unsubstituted carbocyclyl group, a substituted or unsubstituted heterocyclyl group, a substituted or unsubstituted aryl group or a substituted or unsubstituted heteroaryl group, or
R 2 And R 3 Taken together with the carbon atom to which they are attached to form a substituted or unsubstituted 3-8 membered ring;
R 4 and R 5 Each independently hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or substituted or unsubstituted C 1 -C 6 Alkyl, or
R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group;
R 6 absent or hydrogen; and is
Figure FDA0003762336650000012
Represents a single or double bond, wherein
Figure FDA0003762336650000013
When one is a double bond, the other is
Figure FDA0003762336650000014
Is a single bond; when two are
Figure FDA0003762336650000015
When all are single bonds, then R 6 Is hydrogen; and when
Figure FDA0003762336650000016
When one is a double bond, R 6 Is absent.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I-63) is selected from compounds of formula (I-a 63), (I-B63), or (I-C63):
Figure FDA0003762336650000017
Figure FDA0003762336650000021
3. a compound of formula (I-67):
Figure FDA0003762336650000022
or a pharmaceutically acceptable salt thereof, wherein:
R 1 is substituted or unsubstituted alkyl;
R 2 And R 3 Each independently is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or
R 2 And R 3 Taken together with the carbon atoms to which they are attached to form a substituted or unsubstituted 3-8 membered ring;
R 4 and R 5 Each independently is hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or substituted or unsubstituted C 1 -C 6 Alkyl, or
R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group;
R 6 absent or hydrogen; and is
Figure FDA0003762336650000023
Represents a single or double bond, wherein
Figure FDA0003762336650000024
When one is a double bond, the other is
Figure FDA0003762336650000025
Is a single bond; when two are
Figure FDA0003762336650000026
When all are single bonds, then R 6 Is hydrogen; and when
Figure FDA0003762336650000027
When one is a double bond, R 6 Is absent.
4. The compound of claim 3, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I-67) is selected from compounds of formula (I-a 67), (I-B67), or (I-C67):
Figure FDA0003762336650000028
Figure FDA0003762336650000031
5. a compound selected from:
Figure FDA0003762336650000032
Figure FDA0003762336650000041
Figure FDA0003762336650000051
Figure FDA0003762336650000061
Figure FDA0003762336650000071
Figure FDA0003762336650000081
Figure FDA0003762336650000091
Figure FDA0003762336650000101
Figure FDA0003762336650000111
Figure FDA0003762336650000121
6. a pharmaceutical composition comprising a compound of any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
7. Use of a compound according to any one of claims 1 to 5 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to claim 6 in the manufacture of a medicament for the treatment or prevention of a disease as described herein.
8. Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 6, in the manufacture of a medicament for the treatment or prevention of a CNS-related disorder.
9. Use of a compound of formula (A), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for inducing negative allosteric modulation of the NMDA receptor,
Figure FDA0003762336650000131
wherein:
R 1 is hydrogen or substituted or unsubstituted alkyl;
R 2 and R 3 Each independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstitutedCarbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or R 2 And R 3 Taken together with the carbon atoms to which they are attached to form a substituted or unsubstituted 3-8 membered ring;
R 4 and R 5 Each independently is hydrogen, halogen OR-OR C Wherein R is C Is hydrogen or substituted or unsubstituted C 1 -C 6 Alkyl, or
R 4 And R 5 Together with the carbon atom to which they are attached form an oxo group;
R 6 absent or hydrogen;
R G is hydrogen or substituted or unsubstituted alkyl; and is
Figure FDA0003762336650000132
Represents a single or double bond, wherein
Figure FDA0003762336650000133
When one is a double bond, the other is
Figure FDA0003762336650000134
Is a single bond; when two are provided
Figure FDA0003762336650000135
When all are single bonds, then R 6 Is hydrogen; and when
Figure FDA0003762336650000136
When one is a double bond, R 6 Is absent.
10. A method of NMDA modulation comprising assessing NMDA enhancement in a mammalian cell expressing an NMDA receptor using an automated patch clamp system can be used to determine NAM activity of a compound as described below, and PAM activity of a compound can be determined using a whole cell patch clamp system.
CN202210876413.0A 2016-10-18 2017-10-18 Oxysterol and methods of use thereof Pending CN115322238A (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201662409768P 2016-10-18 2016-10-18
US201662409756P 2016-10-18 2016-10-18
US62/409,768 2016-10-18
US62/409,756 2016-10-18
PCT/US2017/057276 WO2018075698A1 (en) 2016-10-18 2017-10-18 Oxysterols and methods of use thereof
CN201780076791.8A CN110072874B (en) 2016-10-18 2017-10-18 Oxysterol and methods of use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201780076791.8A Division CN110072874B (en) 2016-10-18 2017-10-18 Oxysterol and methods of use thereof

Publications (1)

Publication Number Publication Date
CN115322238A true CN115322238A (en) 2022-11-11

Family

ID=60263022

Family Applications (3)

Application Number Title Priority Date Filing Date
CN201780076791.8A Active CN110072874B (en) 2016-10-18 2017-10-18 Oxysterol and methods of use thereof
CN202210425686.3A Pending CN114891056A (en) 2016-10-18 2017-10-18 Oxysterol and methods of use thereof
CN202210876413.0A Pending CN115322238A (en) 2016-10-18 2017-10-18 Oxysterol and methods of use thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CN201780076791.8A Active CN110072874B (en) 2016-10-18 2017-10-18 Oxysterol and methods of use thereof
CN202210425686.3A Pending CN114891056A (en) 2016-10-18 2017-10-18 Oxysterol and methods of use thereof

Country Status (15)

Country Link
US (3) US11111266B2 (en)
EP (2) EP3529256B1 (en)
JP (3) JP2019532079A (en)
KR (2) KR20230051723A (en)
CN (3) CN110072874B (en)
AU (3) AU2017345399B2 (en)
BR (1) BR112019008069A2 (en)
CA (1) CA3041077A1 (en)
ES (1) ES2968140T3 (en)
IL (2) IL293231A (en)
MA (1) MA46565A (en)
MX (2) MX2019004679A (en)
RU (1) RU2019115112A (en)
TW (1) TWI772331B (en)
WO (1) WO2018075698A1 (en)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2968369T3 (en) 2013-03-13 2018-12-17 Sage Therapeutics Inc NEUROACTIVE STEROIDS AND PROCEDURES FOR USE THEREOF
CA2991313C (en) 2015-07-06 2023-12-19 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
MA44526A (en) 2016-04-01 2021-06-02 Sage Therapeutics Inc OXYSTEROLS AND THEIR METHODS OF USE
WO2017193046A1 (en) 2016-05-06 2017-11-09 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
PT3481846T (en) 2016-07-07 2021-08-13 Sage Therapeutics Inc Oxysterols and methods of use thereof
ES2935057T3 (en) 2016-09-30 2023-03-01 Sage Therapeutics Inc C7 substituted oxysterols and these compounds for use as NMDA modulators
KR20230142639A (en) 2016-10-18 2023-10-11 세이지 테라퓨틱스, 인크. Oxysterols and methods of use thereof
US11111266B2 (en) * 2016-10-18 2021-09-07 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US10857163B1 (en) 2019-09-30 2020-12-08 Athenen Therapeutics, Inc. Compositions that preferentially potentiate subtypes of GABAA receptors and methods of use thereof
CN111560045A (en) * 2020-06-23 2020-08-21 江苏佳尔科药业集团股份有限公司 Method for synthesizing lithocholic acid by taking BA as raw material
CN112375117A (en) * 2020-11-14 2021-02-19 湖南科瑞生物制药股份有限公司 Preparation method of lithocholic acid and intermediate thereof
CN113651866B (en) * 2021-08-02 2023-07-04 王涛 Novel method for synthesizing cholesterol by taking 21-hydroxy-20-methyl pregna-4-en-3-one as raw material
WO2023083979A1 (en) * 2021-11-10 2023-05-19 Umecrine Ab 3.beta.-hydroxy, 3.alpha.-ethyl steroids for modulation of the alpha-3 subtype of the gaba-a receptor
WO2023192421A1 (en) 2022-03-30 2023-10-05 Donaldson Company, Inc. System and method for reclaiming solvent
WO2024006537A1 (en) * 2022-07-01 2024-01-04 Intercept Pharmaceuticals, Inc. Methods for ameliorating cognitive impairment using bile acid derivatives

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103958540A (en) * 2011-09-08 2014-07-30 萨奇治疗股份有限公司 Neuroactive steroids, compositions, and uses thereof
CN104136452A (en) * 2011-10-14 2014-11-05 萨奇治疗股份有限公司 3,3 disubstituted 19-nor pregnane compounds, compositions, and uses thereof
CN105164161A (en) * 2013-03-13 2015-12-16 萨奇治疗股份有限公司 Neuroactive steroids, compositions, and uses thereof
CN105246909A (en) * 2013-04-17 2016-01-13 萨奇治疗股份有限公司 19-nor neuroactive steroids and methods of use thereof

Family Cites Families (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2259698A (en) 1938-05-07 1941-10-21 Rare Chemicals Inc Physiologically effective substance and process of preparing same
US2594323A (en) 1948-07-22 1952-04-29 Upjohn Co 24-substituted delta 5-cholene-3, 24-diols
US3079385A (en) 1961-01-24 1963-02-26 Roussel Uclaf Novel process of preparation of polyhydroxylated pregnanes
US3206459A (en) 1962-10-19 1965-09-14 Syntex Corp 10alpha-pregnan-19-ol derivatives
JPS5324071B2 (en) 1974-04-30 1978-07-18
US4071625A (en) 1974-05-13 1978-01-31 Richardson-Merrell Inc. 19-Oxygenated-5α-androstanes for the enhancement of libido
JPS5840960B2 (en) 1976-12-28 1983-09-08 帝人株式会社 Interconversion method between hydroxycholesterol stereoisomers
JPS54163565A (en) 1978-06-09 1979-12-26 Teijin Ltd 24*255epoxyy3beta * 266 dihydroxyychlestoo55ene or protected derivative of hydroxyl group thereof and production
JPS5735597A (en) 1980-08-13 1982-02-26 Teijin Ltd 25,26-epoxy-3beta,24-dihydroxycholest-5-ene or its hydroxyl- protected derivative and their preparation
JPS61254599A (en) 1985-05-07 1986-11-12 Sumitomo Pharmaceut Co Ltd Fluorine derivative of cholesterol
JPS62187485A (en) 1986-02-13 1987-08-15 Teijin Ltd Production of 24,25-epoxycholesterol or such
US5232917A (en) 1987-08-25 1993-08-03 University Of Southern California Methods, compositions, and compounds for allosteric modulation of the GABA receptor by members of the androstane and pregnane series
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
AU698834B2 (en) 1993-05-24 1998-11-12 Purdue Pharma Ltd. Methods and compositions for inducing sleep
IL110309A0 (en) 1993-07-15 1994-10-21 Univ Kentucky Res Found A method of protecting against neuron loss
DE4338316A1 (en) 1993-11-10 1995-05-11 Jenapharm Gmbh New steroids with radicallyophilic substituents, process for their preparation and medicaments containing these compounds
BR9506779A (en) 1994-02-14 1997-10-14 Cocensys Inc Compound pharmaceutical composition method for modulating the gaba-chloride receptor ionophore complex in an animal patient by binding to the neurosteroid site in said complex method to treat or prevent stress or anxiety in an animal patient method to relieve or prevent pms or pnd isonia in an animal patient method for treating or avoiding mood disturbances in an animal patient and method for inducing anesthesia in an animal patient
IL112778A0 (en) 1994-03-04 1995-05-26 Merck & Co Inc Substituted heterocycles, their preparation and pharmaceutical compositions containing them
US5595996A (en) 1994-10-25 1997-01-21 Merck & Co., Inc. 7-substituted 4-aza cholanic acid derivatives and their use
JPH08268917A (en) 1995-03-31 1996-10-15 D D S Kenkyusho:Kk Antitumor agent having high transition to cancer tissue
HUP9901138A3 (en) 1995-06-06 1999-11-29 Cocensys Inc Irvine Neuroactive steroids of the androstane and pregnane series
US6645953B2 (en) 1995-06-23 2003-11-11 Novo Nordisk A/S Meiosis regulating compounds
EP0857173B1 (en) 1995-06-23 2003-11-19 Novo Nordisk A/S Meiosis regulating compounds
EP0840612A1 (en) 1995-07-24 1998-05-13 Trustees Of Boston University Inhibition of nmda receptor activity by pregnenolone sulfate derivatives
US5888996A (en) 1995-07-26 1999-03-30 Trustees Of Boston University Inhibition of NMDA receptor activity and modulation of glutamate-mediated synaptic activity
JPH09328498A (en) 1996-06-10 1997-12-22 Teijin Ltd Production of 24,25-dihydroxycholesterol and its synthetic intermediate
DE19635525A1 (en) 1996-08-20 1998-02-26 Schering Ag New 7-alpha-(xi-aminoalkyl)- oestratriene derivatives
JP2002514621A (en) 1998-05-11 2002-05-21 ノボ ノルディスク アクティーゼルスカブ Substituted guanidines and diaminonitroethenes, their preparation and use
KR20010043558A (en) 1998-05-13 2001-05-25 한센 핀 베네드, 안네 제헤르, 웨이콥 마리안느 Meiosis regulating compounds
US8541600B2 (en) 1998-11-24 2013-09-24 Harbor Therapeutics, Inc. 11-aza, 11-thia and 11-oxa sterol compounds and compositions
DE19917930A1 (en) 1999-04-15 2000-10-19 Schering Ag Treating estrogen deficiency associated disorders, e.g. menopausal problems, osteoporosis, neoplasia or neurodegenerative disease, using new or known ent-steroids having bone-selective estrogenic activity
GB9910934D0 (en) 1999-05-11 1999-07-14 Res Inst Medicine Chem Chemical compounds
CN1098273C (en) 1999-11-12 2003-01-08 中国科学院上海有机化学研究所 High-stereoselectivity synthesization of 24R,25- and 24S, 25-dihydroxysteroid
GB0000228D0 (en) 2000-01-06 2000-03-01 Phytopharm Plc Fluoro substituted sapogenins and their use
GB0019290D0 (en) 2000-08-04 2000-09-27 Symphar Sa Methods for inducing apolipoprotein E secretion
GR1003861B (en) 2000-12-29 2002-04-11 Novel gabaa modulating neurosteroids
GB0107822D0 (en) 2001-03-28 2001-05-23 Phytopharm Plc Sapogenin derivatives their synthesis and use methods based upon their use
US20070197484A1 (en) 2001-05-03 2007-08-23 Ching Song Method of treating disorder related to high cholesterol concentration
WO2003039480A2 (en) 2001-11-08 2003-05-15 The University Of Chicago Method of treating disorder related to high cholesterol concentration
JP2005511713A (en) 2001-12-07 2005-04-28 ザ・リージェンツ・オブ・ザ・ユニバーシティー・オブ・カリフォルニア Treatment for age-related macular degeneration
TWI329016B (en) 2002-03-27 2010-08-21 Phytopharm Plc Therapeutic methods and uses of sapogenins and their derivatives
KR101130212B1 (en) 2002-03-27 2012-04-13 파이토팜 피엘씨 Theraputic methods and uses of SAPOGENIN and their derivatives
US6933312B2 (en) 2002-10-07 2005-08-23 Agouron Pharmaceuticals, Inc. Pyrazole derivatives
WO2004055201A2 (en) 2002-12-13 2004-07-01 Bayer Healthcare Ag Cholesterol 24-hydroxylase (cyp46) as therapeutic target for the treatment of alzheimer's disease
FR2850023B1 (en) 2003-01-17 2007-04-06 Mapreg DRUGS FOR THE NERVOUS SYSTEM
GB0403889D0 (en) 2004-02-21 2004-03-24 Univ Edinburgh Uses of er-beta modulators
WO2006037016A2 (en) 2004-09-27 2006-04-06 The Regents Of The University Of California Novel therapy for treatment of chronic degenerative brain diseases and nervous system injury
CA2643732C (en) 2006-02-27 2012-08-21 The Regents Of The University Of California Oxysterol compounds and the hedgehog pathway
GB0619860D0 (en) 2006-10-06 2006-11-15 Birkeland Innovasjon As Treatment of insulin resistance and disorders associated therewith
US8273747B2 (en) 2006-11-02 2012-09-25 Curis, Inc. Small organic molecule regulators of cell proliferation
ES2662500T3 (en) 2006-11-21 2018-04-06 Asarina Pharma Ab The use of pregnan and androstane steroids for the preparation of a pharmaceutical composition for the treatment of central nervous system disorders
CN101842355A (en) 2007-06-20 2010-09-22 奥斯拜客斯制药有限公司 Substituted n-aryl pyridinones as fibrotic inhibitors
GB0712494D0 (en) 2007-06-27 2007-08-08 Isis Innovation Substrate reduction therapy
CA2703497A1 (en) 2007-11-06 2009-05-14 N.V. Organon A method of hormone suppression in humans
EP2231164A1 (en) 2007-12-03 2010-09-29 The Regents of the University of California Oxysterols for activation of hedgehog signaling, osteoinduction, antiadipogenesis, and wnt signaling
WO2009090063A1 (en) 2008-01-16 2009-07-23 Jado Technologies Gmbh Steroid sapogenin, androstane and triterpenoid sapogenin derivatives for the treatment and prevention of infectious diseases
EA020339B1 (en) 2008-05-09 2014-10-30 Эмори Юниверсити Nmda receptor antagonists for the treatment of neuropsychiatric disorders
WO2010065709A2 (en) 2008-12-03 2010-06-10 Amin Khan Hydroxamic acid derivatives, preparation and therapeutic uses thereof
WO2010075282A1 (en) 2008-12-22 2010-07-01 University Of Washington Molecular inhibitors of the wnt/beta-catenin pathway
US8822462B2 (en) 2009-01-28 2014-09-02 Emory University Subunit selective NMDA receptor potentiators for the treatment of neurological conditions
CN102482315A (en) 2009-07-29 2012-05-30 芝加哥大学 Liver x receptor agonists
WO2011028794A2 (en) 2009-09-01 2011-03-10 Lazarus Therapeutics, Inc. Treatment of huntington's disease with cycloserine and an nmda receptor antagonist
FR2953138B1 (en) 2009-12-02 2015-10-16 Assist Publ Hopitaux Marseille AMINOSTEROIDAL COMPOUNDS FOR LOCAL TOPICAL APPLICATION FOR CUTANEO-MUCOUS DECOLONIZATION OF STAPHYLOCOCCUS AUREUS
WO2011092127A1 (en) 2010-01-26 2011-08-04 Bayer Schering Pharma Aktiengesellschaft 14,17-bridged estratriene derivatives comprising heterocyclic bioisosteres for the phenolic a-ring
KR101692275B1 (en) 2010-02-11 2017-01-04 노오쓰웨스턴 유니버시티 Secondary structure stabilized nmda receptor modulators and uses thereof
US8247436B2 (en) 2010-03-19 2012-08-21 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
US20120035156A1 (en) 2010-08-09 2012-02-09 Daniela Alberati Combination of glyt1 compound with antipsychotics
US8969525B2 (en) 2010-11-09 2015-03-03 Enzo Life Sciences, Inc. Hydroxycholesterol immunoassay
WO2012142039A1 (en) 2011-04-15 2012-10-18 University Of North Dakota Combination of liver x receptor modulator and estrogen receptor modulator for the treatment of age-related diseases
WO2012166617A2 (en) 2011-05-27 2012-12-06 Cytocure Llc Methods, compositions, and kits for the treatment of cancer
JP2014521662A (en) 2011-07-29 2014-08-28 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Novel 17β-heteroaryl substituted steroids as modulators of GABAA receptors
US20150119327A1 (en) 2012-04-25 2015-04-30 The Regents Of The University Of California Drug screening platform for rett syndrome
WO2014025942A1 (en) 2012-08-09 2014-02-13 Emory University Nmda receptor modulators and uses related thereto
US20140149272A1 (en) 2012-08-17 2014-05-29 Trueex Group Llc Interoffice bank offered rate financial product and implementation
JP6542127B2 (en) 2012-12-18 2019-07-10 ワシントン・ユニバーシティWashington University Neuroactive 19-alkoxy-17-substituted steroids, their prodrugs and methods of treatment using the same
EP3309166A3 (en) 2013-01-23 2018-08-22 Sphaera Pharma Pvt. Ltd. 11beta-hydroxy-steroids for use in mitochondria biogenesis and diseases associated with mitochondrial dysfunction or depletion
BR112015018092A2 (en) 2013-01-29 2017-07-18 Naurex Inc spiro-lactam nmda receptor modulators and uses thereof
DK2968369T3 (en) 2013-03-13 2018-12-17 Sage Therapeutics Inc NEUROACTIVE STEROIDS AND PROCEDURES FOR USE THEREOF
US10259840B2 (en) 2014-06-18 2019-04-16 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
EP3166613A4 (en) 2014-07-09 2018-02-21 Duke University Compositions and methods for the repair of myelin
WO2016057713A1 (en) 2014-10-07 2016-04-14 Sage Therapeutics, Inc. Neuroactive compounds and methods of use thereof
CA2991313C (en) 2015-07-06 2023-12-19 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
AU2016289965B2 (en) 2015-07-06 2021-09-09 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
EP3319611B1 (en) 2015-07-06 2021-01-20 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
GB2557875A (en) 2015-09-02 2018-07-04 Univ Swansea Diagnostic methods and kits
MA44526A (en) 2016-04-01 2021-06-02 Sage Therapeutics Inc OXYSTEROLS AND THEIR METHODS OF USE
WO2017193046A1 (en) 2016-05-06 2017-11-09 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
PT3481846T (en) 2016-07-07 2021-08-13 Sage Therapeutics Inc Oxysterols and methods of use thereof
US20190233465A1 (en) 2016-07-11 2019-08-01 Sage Therapeutics, Inc. C7, c12, and c16 substituted neuroactive steroids and their methods of use
ES2935057T3 (en) 2016-09-30 2023-03-01 Sage Therapeutics Inc C7 substituted oxysterols and these compounds for use as NMDA modulators
US11111266B2 (en) 2016-10-18 2021-09-07 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
KR20230142639A (en) 2016-10-18 2023-10-11 세이지 테라퓨틱스, 인크. Oxysterols and methods of use thereof
AU2018234828A1 (en) 2017-03-15 2019-09-19 Modernatx, Inc. Lipid nanoparticle formulation
EP3642217A4 (en) 2017-06-23 2021-06-16 Intercept Pharmaceuticals, Inc. Methods and intermediates for the preparation of bile acid derivatives
EA202091144A1 (en) 2017-11-10 2020-09-16 Маринус Фармасьютикалз, Инк. GANAXOLONE FOR USE IN THE TREATMENT OF HEREDITARY EPILEPTIC DISEASES

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103958540A (en) * 2011-09-08 2014-07-30 萨奇治疗股份有限公司 Neuroactive steroids, compositions, and uses thereof
CN104136452A (en) * 2011-10-14 2014-11-05 萨奇治疗股份有限公司 3,3 disubstituted 19-nor pregnane compounds, compositions, and uses thereof
CN105164161A (en) * 2013-03-13 2015-12-16 萨奇治疗股份有限公司 Neuroactive steroids, compositions, and uses thereof
CN105246909A (en) * 2013-04-17 2016-01-13 萨奇治疗股份有限公司 19-nor neuroactive steroids and methods of use thereof

Also Published As

Publication number Publication date
IL266092A (en) 2019-06-30
CN114891056A (en) 2022-08-12
AU2022200810A1 (en) 2022-02-24
MX2022005160A (en) 2022-06-08
KR102521573B1 (en) 2023-04-14
KR20190065429A (en) 2019-06-11
TW201827057A (en) 2018-08-01
MX2019004679A (en) 2019-10-15
TW202317138A (en) 2023-05-01
CN110072874A (en) 2019-07-30
EP4105223A1 (en) 2022-12-21
AU2017345399A1 (en) 2019-05-09
US20230331768A1 (en) 2023-10-19
ES2968140T3 (en) 2024-05-08
AU2017345399B2 (en) 2022-02-24
EP3529256A1 (en) 2019-08-28
IL293231A (en) 2022-07-01
JP2023063535A (en) 2023-05-09
JP2019532079A (en) 2019-11-07
US11613556B2 (en) 2023-03-28
RU2019115112A3 (en) 2020-12-15
BR112019008069A2 (en) 2019-09-03
IL266092B1 (en) 2023-04-01
US20220024968A1 (en) 2022-01-27
TWI772331B (en) 2022-08-01
AU2022200810B2 (en) 2024-02-01
JP2022033255A (en) 2022-02-28
JP7304444B2 (en) 2023-07-06
EP3529256B1 (en) 2023-08-09
IL266092B2 (en) 2023-08-01
CA3041077A1 (en) 2018-04-26
CN110072874B (en) 2022-08-12
AU2024202773A1 (en) 2024-05-16
US20200123195A1 (en) 2020-04-23
RU2019115112A (en) 2020-11-24
US11111266B2 (en) 2021-09-07
KR20230051723A (en) 2023-04-18
MA46565A (en) 2019-08-28
WO2018075698A1 (en) 2018-04-26

Similar Documents

Publication Publication Date Title
CN110072874B (en) Oxysterol and methods of use thereof
CN110267966B (en) Oxysterol and methods of use thereof
CN109689673B (en) C17, C20 and C21 substituted neuroactive steroids and methods of use thereof
CN115850361A (en) C7 substituted oxysterol and methods of using same as NMDA modulators
TWI836485B (en) Oxysterols and methods of use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination